Abstract
Background
Proton radiation therapy (PRT) and mutant isocitrate dehydrogenase inhibitors (mIDH-inhibitors) are emerging therapies for mIDH lower grade gliomas (LGGs). Despite their substantial theoretical benefits, comparisons with current standards – particularly pertaining to patient-centred outcomes – are limited.
Methods
Through PubMed and Scopus, a search strategy based on keywords focusing on PRT and mIDH-inhibitors was applied on December 3, 2024. Studies in English on at least 20 adult patients (≥ 18 years) with mIDH-LGG grade 2 or 3 and published between January 1, 2011 and August 31, 2024 were included. Review articles were excluded.
Results
Of 6383 identified articles, seven per treatment strategy were included. Overall survival was not reported for mIDH-inhibitors. The lack of high-quality studies comparing PRT to photon radiation therapy precludes conclusions regarding efficacy, effectiveness or even post-PRT radiological manifestations. For the mIDH-inhibitor Vorasidenib (AG-881), the radiological objective response rate was 10.0–42.9%, although lower for contrast-enhancing tumors. Vorasidenib significantly delayed tumor progression (27.7 versus 11.1 months, p < 0.001) and time to next intervention (not reached versus 17.8 months, p < 0.001) when compared to placebo. Adverse events were mostly mild, including elevated liver enzymes (15.6–44.2%) and headache (26.9–46.2%). Only 1/14 studies included assessments related to quality of life (QoL)-domains with inconclusive research on cognitive outcomes.
Conclusion
Studies reporting on patient-centred data including survival, cognition and QoL remain scarce. Larger, comparative prospective studies, preferably randomized controlled trials, with such outcomes are needed to inform clinicians whether the theoretical and radiological benefits can be translated to improved outcomes that matter to patients, i.e. living better and/or longer.
Supplementary information
The online version contains supplementary material available at 10.1007/s00701-025-06612-6.
Keywords: Proton, IDH inhibitor, Vorasidenib, Survival, Quality of life, Glioma
Introduction
Isocitrate dehydrogenase (IDH)-mutant lower grade gliomas (LGGs) are slow growing, infiltrating brain tumors. With new treatments emerging, longer survival of affected patients is anticipated [4, 23, 25, 53]. Therefore, finding effective tumor therapies while simultaneously preserving a good quality of life (QoL) is a priority [51].
Surgical resection is often incomplete and both acute and late side effects are a concern after chemo- and/or radiotherapy [7]. Patients not only face shortened survival but also risk tumor- and treatment-related adverse effects [13, 26, 35, 61]. Nevertheless, current optimism in the field of LGG is present due to emerging therapies, namely proton radiation therapy (PRT) and mutant IDH-inhibitors (mIDH-inhibitors).
PRT holds promise to reduce radiation-related side effects given the beneficial dose-delivery to the healthy brain. Contrary to photons, protons have a determinate range and deposit most of their energy just as they come to a stop in the tissues at the so-called Bragg peak, thereafter, showing a steep dose fall-off. This abrupt stop helps to avoid the widespread low-dose radiation to healthy brain tissue [60].
mIDH-inhibitors such as Vorasidenib can halt the disease owing to their ability to decrease the elevated levels of the oncometabolite D-2-Hydroxyglutarate (2-HG) thereby facilitating cell differentiation, immune cell activation and decreasing tumor proliferation [9, 30, 31, 39].
These emerging therapies, both with a solid theoretical fundament, are increasingly used and reported in patients with mIDH-LGG. Before these approaches are universally or even widely used, it is important to illuminate their impact on aspects that matter to patients. Patients are typically concerned if their survival, cognition or QoL will be impaired. Similarly, patients without access to these treatments may wonder what they are missing out on. In this scoping review, we present the available clinical literature and emphasize the lack of studies demonstrating effects in these patient-centred aspects. Further, we discuss the importance of pushing for evidence beyond surrogate end-points also in slow-growing neoplasms like mIDH-LGGs.
The aim of this scoping review was to investigate the current clinical literature on the demonstrated effects of PRT and mIDH-inhibitors with regards to patient-centred outcomes, including improved survival, QoL or spared cognitive function, in adult patients diagnosed with mIDH-LGG grade 2 or 3.
Methods
Search strategy
Due to the expected limited, heterogenous literature along with the fair recency of these treatments in a group of patients requiring long follow-ups, a scoping review was deemed the most appropriate methodology in order to evaluate the current standing and potentially illuminate gaps in the literature where the need of future research can be directed. This scoping review was conducted, and results reported in alignment with guidelines from Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) [62]. Relevant articles were identified by searching the databases PubMed and Scopus. Assisted by a trained librarian, two search-blocks were formulated based on terms and keywords used in the Medical Subject Headings (MeSH) thesaurus and existing literature on PRT and mIDH-inhibitors (see Supplementary Information 1 for full search-blocks (Online Resource 1)). The search was performed on December 3, 2024.
Eligibility criteria
Retrospective and prospective studies where patients with mIDH-LGG grade 2 or 3 received PRT and/or mIDH-inhibitors were included. Only papers written in English with a sample size of at least 20 adult patients (≥ 18 years old) were considered. Studies with at least 20 total patients of which ≥ 80% of patients had mIDH-tumors of grades 2 or 3 were accepted in studies with mixed populations. The inclusion period was between January 1, 2011, and August 31, 2024, in order to allow some years to pass since the identification of IDH in gliomas [42]. Review articles, studies focusing on laboratory research including animal or studies primarily in pediatric populations were excluded.
Study selection and presentation
Titles and abstracts were initially screened by two independent reviewers (AC and DH). Full-text assessments were made thereafter to ensure eligibility. A third reviewer (ASJ) was consulted when eligibility was doubted. As limited eligible literature was anticipated, no meta-analysis was performed. A PRISMA flowchart (see Fig. 1) was used to demonstrate identification and inclusion of the studies [41]. Through tabulation, the data extracted from included full-text articles was presented in Table 1 for PRT and Table 2 for mIDH-inhibitors.
Fig. 1.
PRISMA flow diagram showing inclusion of studies, IDH = Isocitrate dehydrogenase
Table 1.
Characteristics of studies concerning PRT, n:7
| Number of patients | Main Findings in relation to PRT | ||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|
| Study | Design | Follow-up: Median (range) months | Median Dose, Gy RBE (range) | Total, n | Tumor Grade (G) | IDH-status, n (%) | Aim | Survival | QoL | Toxicities | Radiological manifestations |
| Acharya et al. (2018) [1] | R |
PRT: 19.6 (9.7–31.0) Photon: 34.5 (2.8–109.7) |
PRT: 54.0 (50.4–59.4) Photon: 59.4 (50.4–63.0) |
Total:160 PRT: 37 Photon:123 |
PRT: G2/3: n:19/18 %: 51/49 Photon: G2/3: n: 48/75 %: 39/61 |
PRT: -mIDH: 31(84) Photon: -mIDH: 73 (59) -Unknown: 22 (17) |
Identify risk factors and incidence of developing clinically significant RN in PRT vs. photon treated gliomas | NR | NR | NR |
2-year cumulative incidence for RN: -PRT vs. photon: 18.6% vs. 9.9% - Oligodendroglioma/Astrocytoma: 24.2%/6.2% |
| Dworkin et al. (2018) [14] | R |
Overall: 58.0 (6.0–130.0) PRT + TMZ: 35.0 (NR) PRT only: 64.0 (NR) |
PRT only: 54.0 (45.0–59.4) PRT + concurrent/adjuv-ant TMZ: 56.1 (54.0–60.0) PRT + adjuvant TMZ: 59.4 (54.0–59.4) |
Total: 119 PRT + TMZ: 33 PRT only: 86 |
G2/3: n: 81/38 %: 62/29 |
mIDH: 81 (68) | Investigate whether administering PRT + TMZ vs. only PRT increases risk of PsP development |
-Worse OS and FFP with larger target volumes -Better OS and FFP after 24 months with PsP presence |
NR | NR |
Increased risk for PsP with: -PRT + TMZ (hazard ratio:2.2) -G3 mIDH-tumors (63% vs. 35% in G2 tumors) |
| Eichkorn et al. (2023) [15] | R |
PRT: 5.1 years (7 months-11 years) |
Overall: 54.0 (50.4–60.0) G2: 54.0 (50.4–60.0) G3: 60.0 (54.0–60.0) |
Total: 194 |
G2/3: n:128/66 %:66/34 |
mIDH: 194 (100) | Evaluate the efficacy, safety and long-term clinical and imaging outcomes of PRT in mIDH G2 and G3 gliomas |
5-year OS: G2/3: %: 85/67 5-year PFS: G2/3: %: 60/30 |
NR |
-Acute toxicities, CTCAE < 3a: Fatigue, Headache, Erythema, Focal alopecia -No CTCAE ≥ 3 nor late toxicities |
Overall RICE: 25% of the cohort, of which 31% symptomatic and 35% required therapy RICE related to grade, G2/G3: %: 29/17 RICE: No effect on OS or PFS |
| Ek et al. (2023) [17] | R | Mean follow-up.: PRT: 26.0 (20.0–31.0) Photon: 69.0 (62.0–77.0) |
PRT: 54.0 (54.0–60.0) Photon:59.0 (54.0–64.0) |
PRT: 44 Photon: 98 |
PRT G2/3: n: 29/15 %: 66/34 Photon G2/3: n: 40/58 %: 41/59 |
PRT: -mIDH: 36 (82) Photon: -mIDH: 26 (27) -Unknown: 57 (58) |
Compare treatment outcomes, patient characteristics and survival between PRT and photon therapy |
Decreased PFS and OS with higher average doses Median PFS and OS not reached |
NR |
Acute alopecia CTCAE grade 2: PRT: 15.9% vs. Photon: 5.1% No difference in memory impairment in PRT vs. photon Less acute fatigue CTCAE grade 2 with PRT |
PsP: -Total: n: 21/126 -PRT: n:4, 3% -Photon: n:17, 13% |
| Gómez Vecchio et al. (2024) [19] | P |
PRT: 12.0 (NR) Photon: 12.0 (NR) |
NR |
Total: 51 PRT: 32 Photon: 6 |
G2/3: n: 33/18 %: 65/35 |
mIDH: 51 (100) |
Report on and find predictors of global health status and fatigue changes from preoperative setting to 12 month postoperatively | NR |
PRT: Less unfavourable changes in fatigue No difference in global health status (PRT vs photon) |
NR | NR |
| Qiu et al. (2022) [47] | R | PRT: 21.7 (NR) |
G2: 54.0 (54.0) G3: 60.0 (60.0) G4: 60.0 (54.0–60.0) |
PRT: 52 |
G2/3/4: n: 22/25/5 %: 42/48/10 |
mIDH: 52 (100) | Outcomes of PRT in mIDH gliomas |
PFS: 12/24 months: 97.6%/78.4% OS: 12/24 months: 100%/91% Tumor grade significantly impacted both PFS and OS |
NR |
All acute toxicities: n:40 CTCAE grade 1, Alopecia, n:38 Late toxicities: n:9, CTCAE grade 2 and 3, n:5 including: Dizziness, Fatigue, Memory impairment, Epilepsy |
RN:1.9%; observed in oligodendroglio-ma G3 only |
| Ritterbusch et al. (2021) [50] | R |
PRT: 32.0 (17.0–57.0) Photon: NR |
53.9 (42.0–60.0) | PRT: 57 Photon: 43 |
PRT G2/3: n:21/36 %: 37/63 Photon G2/3: n:12/31 %: 28/72 |
PRT: -mIDH: 49 (86) Photon: -mIDH: 24 (56) -Unknown: 8 (19) |
To distinguish patterns of PsP post-PRT vs PsP after photon therapy |
No difference in OS between: -Patients with/out PsP post-PRT -PRT and photon |
NR |
Asymptomatic PsP: n:5/14 Symptomatic PsP: n:9/14, including vision loss, memory loss |
PsP: -PRT: n:14, 24.6% of which n:9, 64% were symptomatic -Photon: NR |
CTCAE; Common terminology criteria for adverse events, FFP; Freedom from progression, G; Tumor Grade, Gy; Gray, IDH; Isocitrate dehydrogenase, mIDH; mutant- isocitrate dehydrogenase, NR; Not reported, OS; Overall survival, P; Prospective, PFS; Progression Free Survival, PRT; Proton Radiation Therapy, PsP; Pseudoprogression, QoL; Quality of Life, R; Retrospective, RBE; Relative biological effectiveness, RICE; Radiation-induced Contrast Enhancement, RN; Radiation necrosis, TMZ; Temozolomide, vs.; versus
aRates unspecified for each adverse event
Table 2.
Characteristics of studies on mIDH-inhibitors, n:7
| Number of patients | Main Findings in relation to mIDH-inhibitor administration | ||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|
| Study | Design | mIDH-inhibitor | Median treatment duration (range) | Total, n | Tumor Grade (G), | Previous therapy, n (%) | Aim | Survival | QoL | Toxicities | Treatment Response/ORR (%) |
| Cho et al. (2022) [9] | R |
-Ivosidenib (AG-120) -Vorasidenib (AG-881) |
Follow-up: Up to 4 months post-treatment |
Ivosidenib: 18 Vorasidenib: 11 |
G2/3/4: n:19/7/3 %: 66/24/10 |
NR | To investigate MRI changes in gliomas during mIDH-inhibitor treatment at 3–6 weeks and/or 2–4 months after treatment start | NA | NR | NR |
Radiological observations: At 3–6 weeks: Increase in nrCBV and median nrCBV/ADC but not in ADC or FLAIR At 2–4 months: No changes from baseline |
| Mellinghoff et al. (2021) [32] | P, Open-label | Vorasidenib (AG-881) | Enhancing: 3.3 months (0.2–53.6) Non-enhancing: 26.8 months (1.0–50.9) |
Total Glioma: 52 Enhancing: 30 Non-enhancing: 22 |
G2/3/4: n:25/22/4 %: 48/42/8 Unknown: n:1 %: 2 |
Enhancing: -RT: 22 (73) -Systemic: 25 (83) -Only Surgery: 4 (13) Non-enhancing: -RT: 8 (36) -Systemic: 14 (64) -Only surgery: 7 (32) |
Investigate safety and outcomes of Vorasidenib |
Median PFS: -Overall Cohort: 7.5 months -Enhancing: 3.6 months -Non-enhancing: 36.8 months |
NR |
Any CTCAE, n: 52, most common: Headache 46.2% ALT elevation 44.2% CTCAE ≥ 3, n:10 ALT/AST elevation, Fatigue, Nausea, Seizure, Vomiting, Decreased neutrophil count |
Enhancing: ORR (0.0) Non-enhancing: ORR (18.2) |
| Mellinghoff et al. (2023) [30] | RCT, Open-label | Ivosidenib (AG-120) Vorasidenib (AG-881) |
Vorasidenib: 14.3 months (0.9 −22.6) Ivosidenib: 15.1 months (1.8–22.1) |
Vorasidenib: 24 Ivosidenib: 25 |
Vorasidenib: G2/3: n: 22/2 %: 92/8 Ivosidenib: G2/3: n:21/4 %: 84/16 |
Vorasidenib:-Surgery:24 (100)-RT:7 (29)-Systemic:10 (42)Ivosidenib:-Surgery:25 (100)-RT: 7 (28)-Systemic: 14 (56) | To investigate the mechanism of action of Vorasidenib and Ivosidenib | Median PFS not reached in either mIDH-inhibitor | NR |
Vorasidenib: -Any CTCAE, n: 24, most common: Nausea, Headache 41.7%, Diarrhea, Fatigue 29.2% -CTCAE ≥ 3, n:7 Anemia, ALT-elevation, hyperglycemia, hypophosphatemia Ivosidenib: -Any CTCAE, n:25, most common: Headache, Anemia 36.0%, Diarrhea, Seizure 28.0% -CTCAE ≥ 3, n:6, hyponatremia |
Vorasidenib −50mg q.d.: ORR (42.9) −10mg q.d.: ORR (10.0) Ivosidenib −500 mg q.d.: ORR (35.7) −250 mg b.i.d.: ORR (12.5) 2-HG concentration reduction: -Vorasidenib 50 mg q.d.: 92.6% reduction -Ivosidenib 500 mg q.d.: 91.1% reduction |
| Mellinghoff et al. (2023) [31] | RCT, Double-blinded | Vorasidenib (AG-881) |
Median follow-up (Interquartile range): Vorasidenib: 14.0 months (10.1 to 17.9) Placebo: 14.3 months (10.0 to 18.1) |
Vorasidenib: 168 Placebo: 163 |
G2: n:331 %: 100 |
Surgery: 331 (100) | Investigate PFS and time to next intervention in grade 2 tumors receiving Vorasidenib |
Vorasidenib vs placebo: -PFS: 27.7 vs. 11.1 months -Time to next intervention: not reached vs. 17.8 months |
NR |
-Any CTCAE, n:141, most common: ALT elevation 38.9%, Fatigue 32.3% -CTCAE ≥ 3, n:27, ALT/AST/gamma-glutamyl transferase elevation, Fatigue, Diarrhea, Seizure |
Vorasidenib 40 mg q.d.: ORR (10.7) Placebo: ORR (2.5) |
| Natsume et al. (2022) [39] | P, Open-label | Safusidenib (DS-1001) |
Enhancing: 7.3 weeks (0.0–190.0) Non-enhancing 91.2 weeks (15.0–207.0) |
Total: 47 Enhancing: 35 Non-enhancing: 12 |
G2/3/4 n:17/23/7 %36/49/15 |
RT:47 (100)Chemotherapy:38 (81)-TMZ:35 (75)-Nimusutine14 (30)-Bevacizumab 7 (15) |
To determine the safety, efficacy and pharmacology of Safusidenib/ DS-1001 |
Median PFS Non-enhancing vs. Enhancing: not reached vs.10.4 weeks |
NR |
-All CTCAE, n:45, most common: Skin hyperpigmentation 53.2% Diarrhea 46.8% -CTCAE 3, n: 20, Diarrhea, Arthralgia, headache, decreased neutrophil count, ALT/AST elevation |
-Significantly lower 2-HG in Safusidenib (DS-1001) tumor samples -Enhancing: ORR (17.1) (CR: n: 2, 5.7%, PR: n: 4, 11.4%) -Non-enhancing: ORR (33.3) (PR: n: 1, 8.3%, mR: n: 3, 25.0%) |
| Peters et al. (2023) [43] | R | Ivosidenib (AG-120) | 43.7 weeks (5.5- 74.1) |
Total: 30 Enhancing: 8 Non-enhancing: 22 |
G2/3/4: n:21/8/1 %:70/27/3 |
RT: 8 (27)TMZ: 18(60)mIDH-peptide vaccine: 2 (7)Bevacizumab1 (3) | To investigate the outcomes of Ivosidenib treatment | Deaths: n:2 (of total 30) | NR |
CTCAE 1, n:23, most common: Diarrhea 26.7%, Elevated creatine kinase 33.3% |
Enhancing: ORR (0.0) Non-enhancing: ORR (36.3) |
| Wick et al. (2021) [65] | P | BAY1436032 | NR |
Total glioma:39 Enhancing: 33 Non-enhancing: 2 Unknown: 4 Dose expansion glioma: 25 Dose escalation glioma: 14 |
NR | Dose expansion: -RT: 23 (92)Dose escalation:NR separately | To investigate the safety profile and pharmacology of BAY1436032 in mIDH solid tumors | PFS rate at 3 months: 0.31 | NR | NA due to analysis with other tumor types | ORR: (11) (CR: n:1, 3.0%, PR: n:3, 9.0%) |
2-HG; 2-hydroxyglutarate, ADC; Apparent diffusion coefficient, ALT; alanine aminotransferase, AST; aspartate aminotransferase, b.i.d.; bis in die, twice a day, CR; Complete Response, CTCAE; Common terminology criteria for adverse events, FLAIR; Fluid-attenuated inversion recovery, G; Tumor grade, mIDH; mutant-isocitrate dehydrogenase, mR; minor Response, MRI; Magnetic Resonance Imaging, NA; Not Applicable, NR; Not reported, nrCBV; normalized relative cerebral blood volume, ORR; Objective response rate, P; Prospective, PFS; Progression Free Survival, PR; Partial Response, q.d.; quaque die, once a day, QoL; Quality of Life, R; Retrospective, RCT; Randomized controlled trial, RT; Radiotherapy, TMZ; Temozolomide, vs.; versus
Results
Our search identified a total of 6383 articles from PubMed and Scopus with gradual assessments leading to the final inclusion of seven articles per treatment (see Fig. 1).
Proton radiation therapy
Across all included studies, a total of 535 patients received PRT [1, 14, 15, 17, 19, 47, 50]. All but one article had a retrospective study design, and the single prospective study focused on longitudinal changes in patient-reported outcomes where radiation therapy was a covariate [19]. No eligible randomized controlled trial (RCT) was identified. Median follow-up ranged between 12–64 months and median PRT-dose prescribed was 54–60 Gy RBE across studies. For a detailed overview, see Table 1.
Survival
While median overall survival (OS) was not reached in two studies, [15, 17], one study reported a progression free survival (PFS) and OS at 24-months after PRT to 78.4% and 91.0%, respectively [47]. In the same study, tumor grade impacted OS and PFS the most as shown by univariate (OS: p = 0.002, PFS: p = 0.004) and multivariate analysis (OS: p = 0.024, PFS: p = 0.022) [47]. Another study also indicated the importance of tumor grade, with the 5-year OS in grade 2/3 tumors being 85.0%/67.0%, respectively (p = 0.0088) [15]. Similarly, the 5-year PFS in grade 2 and 3 tumors were 60.0% and 30.0%, respectively (p < 0.0001) [15].
Two other studies compared survival in patients receiving PRT versus photon radiation therapy [17, 50]. None of the studies showed differences in OS between radiation modalities. Moreover, the proportion of mIDH-tumors in the photon therapy groups in both studies did not satisfy this review’s eligibility criteria.
Toxicities
Four studies included reports on adverse events [15, 17, 47, 50]. Toxicities mostly occurred early in the post-PRT period and were mild, grading 1–2 on the common terminology criteria for adverse events (CTCAE) (see Table 1). The reporting on specific mild toxicities varied between 1.9% and 73.1%. The most common toxicity was alopecia (15.9–73.1%), but adverse events also included headache, erythema and fatigue. Late, severe toxicities were reported by two studies [47, 50].
The prospective study by Gómez Vecchio et al. indicated a lower proportion of PRT patients experiencing unfavorable changes in fatigue in comparison to photon therapy (66.7% vs. 95.7%, p = 0.03) [19]. No differences related to changes in global health status as a QoL-construct were noted (p = 0.61) [19]. Another study reported a higher rate of fatigue in photon therapy than PRT up to 3 months posttreatment (p = 0.016) thereafter stabilizing in both modalities. The same study reported no difference in subjective memory impairment between radiation modalities [17]. However, the photon therapy group here was not molecularly defined to the same extent, precluding valid conclusions. Other studies reported individual cases developing impaired memory [47, 50].
Radiological manifestations
Pseudoprogression (PsP) was discussed in three studies [14, 17, 50]. In one study, no difference was found in PsP-rate between patients treated with photons versus protons (p = 0.38) [17]. Another study showed PsP in 43.6% (52/119) of PRT-patients with an increased risk of PsP-development in grade 3 versus grade 2 tumors (p = 0.003) [14]. However, more grade 2 tumors here had unknown IDH-status. Therefore, caution should be exerted when interpreting these findings [14]. Ritterbusch et al. observed unique imaging patterns of PsP following PRT. There, PsP developed in 24.6% of patients and PsP-lesions were characterized by being small (< 1 cm), transient, multifocal and patchy with a location at the Bragg peak. The mean time to PsP-occurrence following PRT was 15.4 months while photon-related PsP occurred within 3 months [50]. None of the studies showed association between PsP and tumor subtypes [14, 17, 50]. Finally, Eichkorn et al. assessed radiation induced contrast enhancement (RICE), developing in 25.3% of their overall cohort, and in 29.7% of grade 2 versus 16.7% of grade 3 patients (p = 0.11) [15]. The presence of RICE did not influence OS (p = 0.15) nor PFS (p = 0.67) [15]. However, astrocytomas were associated with earlier RICE development in comparison to oligodendrogliomas (p = 0.047) and RICE risk increased independently with older age (p = 0.017) [15].
Despite the large proportion of IDH-wildtype tumors in the photon therapy group, Acharya et al. reported the 2-year cumulative incidence for radiation necrosis (RN) following PRT to be 18.6% compared with 9.9% after photon therapy (p = 0.16) [1]. Further, the incidence of RN was higher in oligodendrogliomas compared to astrocytomas (p = 0.01) with no difference in radiation modality observed (p = 0.68) [1]. Another study reported a single case of severe RN (CTCAE grade 3) in their cohort, observed in a patient with oligodendroglioma grade 3 treated with PRT [47].
Mutant IDH-inhibitors
Out of seven studies on mIDH-inhibitors, two had a retrospective design, [9, 43], while the rest were prospective studies. A total of 73 patients received Ivosidenib (AG-120), 255 received Vorasidenib (AG-881), 47 received Safusidenib (DS-1001), and 39 received BAY1436032. For a detailed overview, see Table 2.
Progression free survival
None of the seven articles reported on OS while six studies reported PFS [9, 30–32, 39, 65]. In an RCT examining Ivosidenib in non-enhancing LGG, median PFS was not reached after a median postoperative treatment duration of 15.1 months [30]. Another study reported two deaths (of 30 patients in total) after a median time on Ivosidenib of 43.7 weeks [43].
With Vorasidenib, a median PFS of 7.5 months was seen in a broad mIDH glioma cohort, with longer PFS in non-enhancing versus enhancing tumors (36.8 vs. 3.6 months, respectively) [32]. This difference in PFS between enhancing and non-enhancing lesions was seen also with Safusidenib (DS-1001). There, median PFS was 10.4 weeks in enhancing tumors while median PFS was not reached in non-enhancing gliomas after a median treatment duration of 7.3 and 91.2 weeks, respectively [39]. At 3 months, BAY1436032 showed a PFS of 31.0% [65]. The study by Cho et al. included joint data on Ivosidenib and Vorasidenib [9]. A shorter PFS was associated with: 1) continuously decreasing diffusion coefficient on imaging, 2) Fluid-attenuated inversion recovery (FLAIR) volume increase > 4cm3 and 3) elevated perfusion values at 2–4 months posttreatment. However, the authors stress not to interpret this as prognostic effects of mIDH-inhibitors, rather as preliminary radiographic observations [9].
Lastly, in the INDIGO double-blinded RCT, Vorasidenib showed significantly prolonged median PFS versus placebo (27.7 vs. 11.1 months; p < 0.001) in grade 2 non-enhancing tumors after a median follow-up of 14 months [31]. Similarly, Vorasidenib improved median time to next intervention (not reached vs. 17.8 months, p < 0.001) [31].
Toxicities
Mild toxicities (CTCAE grade < 3) dominated in patients treated with mIDH-inhibitors (see Table 2). With Vorasidenib, headache, nausea and liver enzyme elevation were common. Other infrequent toxicities included diarrhea, constipation and electrolyte and/or glucose disturbances [30–32]. Fatigue was reported at rates around 30% and subjective memory impairment at 16.7% [9, 30–32]. Ivosidenib reported analogous toxicity rates to Vorasidenib, but more electrolyte and/or glucose disturbances and anemia were reported with Ivosidenib. Seizure frequency increased in 13.3% of patients and fatigue constituted 12.0% of all toxicities of Ivosidenib [30, 43].
Safusidenib (DS-1001) exclusively caused frequent dermatological complications in up to 53.2% of patients, alopecia/arthralgia in 27.7% and backaches in 21.3%. Diarrhea was more common while rates of headache and nausea were comparable to other mIDH-inhibitors. Limited electrolyte disturbances or elevation of liver enzymes were noted [39]. Toxicities of BAY1436032 were analysed alongside other tumor types and included diarrhea, nausea, fatigue and liver enzyme elevation [65].
Therapeutic response
Neither Vorasidenib nor Ivosidenib showed complete responses (CR). In non-enhancing tumors, the highest objective response rate (ORR) was noted following 50 mg Vorasidenib (ORR 42.9%) [30]. The ORR decreased to 10.0% when administering 10 mg Vorasidenib, indicating a dose–response effect. Another Vorasidenib trial showed an ORR of 18.2% in non-enhancing tumors, with a partial response of 4.5% occurring at 50 mg once daily and minor response of 13.6% at 200 mg once daily [32]. No radiographic response was noted in enhancing tumors [32]. The INDIGO trial reported an ORR of 10.7% (vs. 2.5% in placebo) with the administration of 40 mg Vorasidenib once daily [31]. Thus, the range of ORR with Vorasidenib across studies was 10.0–42.9%, at least partially explained by administered doses. Lastly, a 92.6% reduction in 2-HG concentrations in resected tumor tissues was noted in addition to decreased tumor proliferation, more activated immune cells and suppressed genes related to cell-cycle progression. Of note, these analyses were performed in patients who received Vorasidenib 50 mg once daily for 28 (+ 7) days, up until surgery [30].
Ivosidenib also showed better response in non-enhancing versus enhancing tumors (ORR 36.3% vs. 0.0%) [43]. With the administration of 500 mg Ivosidenib once daily, an ORR of 35.7% was reported. The ORR decreased almost threefold with the administration of 250 mg Ivosidenib twice a day instead (ORR 12.5%) [30]. The concentrations of 2-HG in resected tumor tissues were consequently decreased by 91.1% following the administration of 500 mg Ivosidenib once daily for 28 (+ 7) days, up until surgery [30].
Among all mIDH-inhibitors, Safusidenib (DS-1001) reported the highest CR of 5.7% and exclusively in enhancing tumors. Following Safusidenib (DS-1001) administration twice per day in 21-day cycles, the ORR observed was 33.3% in non-enhancing tumors versus 17.1% in enhancing tumors. Further, tumor samples of patients still receiving Safusidenib (DS-1001) but underwent surgery upon progression showed lower 2-HG concentrations after treatment [39]. BAY1436032 specifically targeted enhancing tumors and demonstrated CR in 3.0% of cases [65].
Discussion
In this scoping review, we demonstrate promising results on surrogate outcomes following both PRT and mIDH-inhibitors. Although the studies involving mIDH-inhibitors were more often prospective and randomized than in PRT, some of the same shortcomings exist. Thus far, no solid evidence exists with regards to patient-centred outcomes such as improved survival, QoL or spared cognitive function following these therapies especially in relation to other treatment options.
Patient-centred outcomes
To date, only PFS and time to next intervention has been reported for mIDH-inhibitors. Substantial data on OS, QoL and cognition is lacking. Survival comparisons between PRT and photon therapy were limited by the low proportion of mIDH-tumors in the photon groups, often included at an earlier time-point [17, 50]. It is yet to be explored what influences neurocognition in mIDH-LGG; whether it is the disease itself, the treatments applied or a combination of both [11, 58, 59, 67]. Dosimetric studies assessing PRT showed a comparable target volume to photon therapy and delivery of lower radiation doses to organs at risk [2, 22, 68]. This spared critical neurological structures, including areas of neurogenesis (subventricular zone and hippocampus), thereby showing potential in decreasing long term sequelae and improving QoL [2, 22]. Only one included study associated PRT with less unfavourable changes in fatigue affecting QoL [19]. Other included studies reported fatigue, memory impairment and other adverse events without elaborating on specific QoL-domains. Studies not included in this review, assessed QoL with PRT and showed development of neuroendocrine deficiencies with patients retaining cognitive function overtime [55, 57]. In pediatric populations, PRT was associated with less cognitive impairment [20, 33, 38, 45].
Surrogate end-points
Surrogate end-points applied in this context included PFS and dosimetric advantages. In recent years, cancer drugs showing only a PFS benefit were approved by the Food and Drug Administration [44]. Due to limited therapies for mIDH-LGG, the application of surrogate end-points might accelerate the availability of novel treatments in the market. However, the long-term benefits of these treatments remain unknown and surrogate end-points do not necessarily reflect on outcomes that matter to patients [52, 64].
It remains unclear whether an improved PFS translates into improved survival, QoL or cognition. Deterioration in clinical symptoms does not always correlate with pre-defined radiological progression criteria [34, 52]. A comparable case involved Bevacizumab in glioblastoma-treatment which, despite a beneficial PFS, did not ultimately prolong OS nor improve QoL but was more frequently associated with serious toxicities [8, 12, 18]. Similarly, another trial comparing early versus delayed radiotherapy in LGG showed a PFS benefit with early post-surgical radiotherapy that could not, however, be translated to improved OS [63]. Even in grade 2 mIDH-glioma, predicting OS or post-progression survival based upon PFS has proven difficult [52]. The dosimetric advantages of PRT may be offset by post-PRT manifestations such as PsP, RICE or RN, especially as these per se may be difficult to differentiate from true tumor progression, thereby leading to unnecessary, and sometimes deleterious, interventions due to misdiagnosis [1, 16, 24, 40, 59]. Perhaps assessing clinical outcomes could better guide treatment decision-making [52].
Post-PRT radiological manifestations
Contrast-enhancing brain lesions, RN, PsP and RICE were apparent in several included studies [1, 14, 15, 17, 47, 50]. However, the heterogeneity of the results, populations and terminology used to describe these manifestations complicates comparisons and prevents conclusions from being drawn [17, 27, 29]. These inconsistencies in reporting were even apparent in other studies not included in this review.
In similarity to the included study by Acharya et al. [1], another study, not included in this review according to the eligibility criteria, reported a higher proportion of tissue necrosis following PRT compared to photon therapy (18% vs. 6% respectively) [66]. However, consistent with findings from another included study [17], Bronk et al. observed no difference in PsP rate between radiation modalities [6].
Among the included studies, PsP was more frequently observed in grade 3 tumors while RICE rate was higher in grade 2 tumors [14, 15]. PsP was not associated with tumor subtypes [14, 17, 50]. However, astrocytomas showed earlier RICE development, while oligodendroglioma patients -regardless of treatment modality- expressed a higher incidence of RN with one case of severe RN following PRT in grade 3 oligodendroglioma [1, 15, 47]. Despite not meeting this review’s eligibility criteria, Bronk et al. showed earlier occurrence of PsP in oligodendroglioma following PRT and no correlation between PsP rate and tumor grade [6]. In another photon-dominated cohort, all RN cases following PRT occurred in patients with oligodendroglioma [3]. However, too few patients were present in both studies to be able to conclude on this much debated topic [3, 6]. Older age was shown to be an independent risk factor for RICE in one included study [15]. While oligodendrogliomas usually present at higher median ages [25], another study showed slightly higher RICE rate in astrocytomas (50% vs. 46% in oligodendrogliomas) [16]. Hence, inconclusive research is currently present regarding associations between tumor subtype and post-PRT manifestations.
Limitations
Our search was dominated by single-institutional, retrospective, uncontrolled studies with small sample sizes, mixed IDH-mutational status and short follow-ups. Nonetheless, this reflects the present state of evidence. The latest 2021 World Health Organization classification of tumors was not always applied [28]. For PRT, selection was biased to younger patients, less pre-radiation cognitive impairment and lower tumor grades with lower administered doses in accordance with the current guidelines in order to minimize neurocognitive effects [1, 17, 19]. Further, inconsistent definitions and different nomenclature used to describe post-PRT radiological manifestations prevents current comparisons between proton studies further complicating the clinical application of these manifestations and the gathering of results on common ground [17, 27, 29].
For mIDH-inhibitors, prior therapies and short follow-ups complicate effects on survival, while placebo cross-over in the INDIGO-trial will complicate long term survival analysis. In the INDIGO trial, patients receiving placebo could cross-over to Vorasidenib upon progression. Given the aim to delay radio-chemotherapy, the threshold to receive Vorasidenib as second-line therapy for placebo patients was probably lower than to receive radio-chemotherapy, which was the option in the Vorasidenib arm of this trial [31]. Further, the INDIGO trial compared treatment-naïve patients receiving Vorasidenib monotherapy to placebo [31]. This may limit comparisons to the current standards of LGG treatment. On the other hand, it provides the opportunity for studying a watch-and-wait strategy that is applied in selected centres following surgical resection and compare it with active treatment with presumably less neurotoxic effects [5, 13, 31, 32].
Despite the current caveats, the cumulative evidence suggests that lower-grade, non-enhancing tumors are responsive to mIDH-inhibitors with regards to progression and metabolic alterations such as reduced 2-HG concentrations. For mIDH-LGGs having undergone malignant transformation, studies incorporating combination therapies and trials in enhancing tumors are coveted. Previous studies in acute myeloid leukemia showed significant advantage when mIDH-inhibitors were combined with the hypomethylating agent, Azacitidine [10, 37]. Interestingly, Safusidenib (DS-1001) and BAY1436032 produced CR in enhancing tumors, even providing preliminary prospects for systematic targeted therapy in mIDH gliomas of higher grade as well [39, 65].
Future directions
Assessing OS in mIDH-LGGs might take decades, but relying on surrogate end-points is only a short-term solution. Given the long-expected survival of patients, often undergoing multiple interventions over time, therapies directed at the early stages of disease should prove long-term benefits [52]. Prospective studies with longer follow-ups examining patient-centred domains are, therefore, highly encouraged. Awaiting us in the field of PRT in mIDH-glioma are three prospective, randomized trials; PRO-GLIO, NRG-BN005, and APPROACH; leveraging on survival and cognitive outcomes [21, 23, 56]. Additionally, the production of consensus-based definitions may ease the reporting, analysis and application of post-PRT radiological changes in future studies, a work that has already been started by the European Particle Therapy Network [27]. The long-term OS, cognitive and QoL analyses from the INDIGO trial and further trials on Safusidenib (DS-1001) and Vorasidenib through the VIGOR trial are even forecasted [31, 46, 54]. Finally, to facilitate the holistic reporting and analysis of trial outcomes aligning with patient concerns and priorities, the development of core outcome sets in future trials concerning these treatments is worth considering. So far for adult gliomas, the COBra-study developed a core outcome set where some of the main outcome domains included survival, health-related QoL and cognitive function [36, 48, 49].
Conclusion
PRT and mIDH-inhibitors are two therapies promising precision in targeting mIDH-LGG. Current reports lack profound assessments of patient-centred outcomes despite their prognostic importance in the young, affected patient group. Prospective, patient-centred studies, preferably RCTs, with longer follow-ups and larger populations, are strongly encouraged to aid treatment decisions.
Supplementary information
Below is the link to the electronic supplementary material.
(PDF 97.0 KB)
Acknowledgements
The authors would like to thank Linda Hammarbäck (LH) from the Gothenburg University Library for her help and advice on data search as well as creation of the search blocks applied in this scoping review.
Abbreviations
- 2-HG
D-2-Hydroxyglutarate
- CR
Complete Response
- CTCAE
Common terminology criteria for adverse events
- IDH
Isocitrate-dehydrogenase
- LGGs
Lower grade gliomas
- mIDH-inhibitors
Mutant isocitrate-dehydrogenase-inhibitors
- ORR
Objective response rate
- OS
Overall survival
- PFS
Progression free survival
- PRT
Proton radiation therapy
- PsP
Pseudoprogression
- QoL
Quality of life
- RCT
Randomized controlled trial
- RICE
Radiation induced contrast enhancement
- RN
Radiation necrosis
Author contributions
A.S.J. conceptualized and designed the framework of this scoping review. A.C. and D.H conducted the literature search and screening of titles and abstracts. A.C., A.S.J. and D.H. were involved in the eligibility assessment of full-texts. D.H. drafted the manuscript. A.C., A. M., A.S.J., M.B. and P.B. critically revised the manuscript. All authors reviewed and approved the final version of the manuscript for publication.
Funding
Open access funding provided by University of Gothenburg. A.S.J. received grants from the Swedish state under the agreement between the Swedish government and the county councils, the ALF-agreement (ALFGBG-1006089). M.B. received grants from Jubileumsklinikens Cancerfond, the Swedish Society of Medicine and the Gothenburg Society of Medicine. The funders had no role in the preparation of this scoping review.
Data availability
No datasets were generated or analysed during the current study.
Declarations
Ethical approval
No ethical approval from an ethics committee or institutional review board was required as no access to sensitive patient information nor any direct involvement of human/animal participants was included in this review.
Consent to participate
Not applicable.
Consent to publish
Not applicable.
Competing interests
Author P.B. declares honoraria for lectures, consultation or advisory board participation from Servier, Ekspertpanelet and Moloklinikken.
The remaining authors have no conflicts of interest to declare that are relevant to the content of this article.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Acharya S, Robinson CG, Michalski JM, Mullen D, DeWees TA, Campian JL, Chundury A, Bottani B, Hallahan DE, Bradley JD, Huang J (2018) Association of 1p/19q codeletion and radiation necrosis in adult cranial Gliomas after proton or photon therapy. Int J Radiat Oncol Biol Phys 101:334–343. 10.1016/j.ijrobp.2018.01.099 [DOI] [PubMed] [Google Scholar]
- 2.Adeberg S, Harrabi SB, Bougatf N, Verma V, Windisch P, Bernhardt D, Combs SE, Herfarth K, Debus J, Rieken S (2018) Dosimetric Comparison of Proton Radiation Therapy, Volumetric Modulated Arc Therapy, and Three-Dimensional Conformal Radiotherapy Based on Intracranial Tumor Location. Cancers (Basel) 10:401 10.3390/cancers10110401 [DOI] [PMC free article] [PubMed]
- 3.Ahmad H, Martin D, Patel SH, Donahue J, Lopes B, Purow B, Schiff D, Fadul CE (2019) Oligodendroglioma confers higher risk of radiation necrosis. J Neurooncol 145:309–319. 10.1007/s11060-019-03297-7 [DOI] [PubMed] [Google Scholar]
- 4.Baek C, Laurenge A, Touat M (2024) Advances in the treatment of IDH-mutant gliomas. Curr Opin Neurol 37:708–716. 10.1097/wco.0000000000001316 [DOI] [PubMed] [Google Scholar]
- 5.Bhatia A, Moreno R, Reiner AS, Nandakumar S, Walch HS, Thomas TM, Nicklin PJ, Choi Y, Skakodub A, Malani R, Prabhakaran V, Tiwari P, Diaz M, Panageas KS, Mellinghoff IK, Bale TA, Young RJ (2024) Tumor Volume Growth Rates and Doubling Times during Active Surveillance of IDH-mutant Low-Grade Glioma. Clin Cancer Res 30:106–115. 10.1158/1078-0432.Ccr-23-1180 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bronk JK, Guha-Thakurta N, Allen PK, Mahajan A, Grosshans DR, McGovern SL (2018) Analysis of pseudoprogression after proton or photon therapy of 99 patients with low grade and anaplastic glioma. Clin Transl Radiat Oncol 9:30–34. 10.1016/j.ctro.2018.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Capelle L, Fontaine D, Mandonnet E, Taillandier L, Golmard JL, Bauchet L, Pallud J, Peruzzi P, Baron MH, Kujas M, Guyotat J, Guillevin R, Frenay M, Taillibert S, Colin P, Rigau V, Vandenbos F, Pinelli C, Duffau H (2013) Spontaneous and therapeutic prognostic factors in adult hemispheric World Health Organization grade II Gliomas: a series of 1097 cases: clinical article. J Neurosurg 118:1157–1168. 10.3171/2013.1.Jns121 [DOI] [PubMed] [Google Scholar]
- 8.Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D, Brandes AA, Hilton M, Abrey L, Cloughesy T (2014) Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med 370:709–722. 10.1056/NEJMoa1308345 [DOI] [PubMed] [Google Scholar]
- 9.Cho NS, Hagiwara A, Eldred BSC, Raymond C, Wang C, Sanvito F, Lai A, Nghiemphu P, Salamon N, Steelman L, Hassan I, Cloughesy TF, Ellingson BM (2022) Early volumetric, perfusion, and diffusion MRI changes after mutant Isocitrate Dehydrogenase (IDH) inhibitor treatment in IDH1-mutant Gliomas. Neurooncol Adv 4:vdac124. 10.1093/noajnl/vdac124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.DiNardo CD, Schuh AC, Stein EM, Montesinos P, Wei AH, de Botton S, Zeidan AM, Fathi AT, Kantarjian HM, Bennett JM, Frattini MG, Martin-Regueira P, Lersch F, Gong J, Hasan M, Vyas P, Döhner H (2021) Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): a single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol 22:1597–1608. 10.1016/s1470-2045(21)00494-0 [DOI] [PubMed] [Google Scholar]
- 11.Dirven L, Reijneveld JC, Taphoorn MJB, Coens C, El-Badawy SA, Tzuk-Shina T, Bravo-Marques J, Back M, Stalpers LJA, Stupp R, Baumert BG, Seidel C (2019) Impact of Radiation Target Volume on Health-Related Quality of Life in Patients With Low-Grade Glioma in the 2-Year Period Post Treatment: A Secondary Analysis of the EORTC 22033–26033. Int J Radiat Oncol Biol Phys 104:90–100. 10.1016/j.ijrobp.2019.01.003 [DOI] [PubMed] [Google Scholar]
- 12.Dirven L, van den Bent MJ, Bottomley A, van der Meer N, van der Holt B, Vos MJ, Walenkamp AM, Beerepoot LV, Hanse MC, Reijneveld JC, Otten A, de Vos FY, Smits M, Bromberg JE, Taal W, Taphoorn MJ (2015) The impact of bevacizumab on health-related quality of life in patients treated for recurrent glioblastoma: results of the randomised controlled phase 2 BELOB trial. Eur J Cancer 51:1321–1330. 10.1016/j.ejca.2015.03.025 [DOI] [PubMed] [Google Scholar]
- 13.Douw L, Klein M, Fagel SS, van den Heuvel J, Taphoorn MJ, Aaronson NK, Postma TJ, Vandertop WP, Mooij JJ, Boerman RH, Beute GN, Sluimer JD, Slotman BJ, Reijneveld JC, Heimans JJ (2009) Cognitive and radiological effects of radiotherapy in patients with low-grade glioma: long-term follow-up. Lancet Neurol 8:810–818. 10.1016/s1474-4422(09)70204-2 [DOI] [PubMed] [Google Scholar]
- 14.Dworkin M, Mehan W, Niemierko A, Kamran SC, Lamba N, Dietrich J, Martinez-Lage M, Oh KS, Batchelor TT, Wen PY, Loeffler JS, Shih HA (2019) Increase of pseudoprogression and other treatment related effects in low-grade glioma patients treated with proton radiation and temozolomide. J Neurooncol 142:69–77. 10.1007/s11060-018-03063-1 [DOI] [PubMed] [Google Scholar]
- 15.Eichkorn T, Lischalk JW, Hörner-Rieber J, Deng M, Meixner E, Krämer A, Hoegen P, Sandrini E, Regnery S, Held T, Harrabi S, Jungk C, Herfarth K, Debus J, König L (2023) Analysis of safety and efficacy of proton radiotherapy for IDH-mutated Glioma WHO grade 2 and 3. J Neurooncol 162:489–501. 10.1007/s11060-022-04217-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Eichkorn T, Lischalk JW, Sandrini E, Meixner E, Regnery S, Held T, Bauer J, Bahn E, Harrabi S, Hörner-Rieber J, Herfarth K, Debus J, König L (2022) Iatrogenic influence on prognosis of radiation-induced contrast enhancements in patients with glioma WHO 1–3 following photon and proton radiotherapy. Radiother Oncol 175:133–143. 10.1016/j.radonc.2022.08.025 [DOI] [PubMed] [Google Scholar]
- 17.Ek H, Fagerström Kristensen I, Stenberg L, Kinhult S, Benedek H, Ek S, Engelholm SA, Engelholm S, Munck Af Rosenschöld P (2023) Transitioning from conventional photon therapy to proton therapy for primary brain tumors. Acta Oncol 62:391–399. 10.1080/0284186x.2023.2200150 [DOI] [PubMed] [Google Scholar]
- 18.Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M, Jeraj R, Brown PD, Jaeckle KA, Schiff D, Stieber VW, Brachman DG, Werner-Wasik M, Tremont-Lukats IW, Sulman EP, Aldape KD, Curran WJ Jr, Mehta MP (2014) A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 370:699–708. 10.1056/NEJMoa1308573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gómez Vecchio T, Rydén I, Ozanne A, Blomstrand M, Carstam L, Smits A, Jakola AS (2024) Global health status and fatigue score in isocitrate dehydrogenase-mutant diffuse glioma grades 2 and 3: A longitudinal population-based study from surgery to 12-month follow-up. Neurooncol Pract 11:347–357. 10.1093/nop/npae017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Greenberger BA, Pulsifer MB, Ebb DH, MacDonald SM, Jones RM, Butler WE, Huang MS, Marcus KJ, Oberg JA, Tarbell NJ, Yock TI (2014) Clinical outcomes and late endocrine, neurocognitive, and visual profiles of proton radiation for pediatric low-grade gliomas. Int J Radiat Oncol Biol Phys 89:1060–1068. 10.1016/j.ijrobp.2014.04.053 [DOI] [PubMed] [Google Scholar]
- 21.Grosshans D, NRG Oncology, National Cancer Institute (NCI) (2017) Proton Beam or Intensity-Modulated Radiation Therapy in Preserving Brain Function in Patients With IDH Mutant Grade II or III Glioma. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT03180502. Accessed 27 Feb 2025
- 22.Harrabi SB, Bougatf N, Mohr A, Haberer T, Herfarth K, Combs SE, Debus J, Adeberg S (2016) Dosimetric advantages of proton therapy over conventional radiotherapy with photons in young patients and adults with low-grade glioma. Strahlenther Onkol 192:759–769. 10.1007/s00066-016-1005-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Heggebø LC, Borgen IMH, Rylander H, Kiserud C, Nordenmark TH, Hellebust TP, Evensen ME, Gustavsson M, Ramberg C, Sprauten M, Magelssen H, Blakstad H, Moorthy J, Andersson K, Raunert I, Henry T, Moe C, Granlund C, Goplen D, Brekke J, Johannessen TA, Solheim TS, Marienhagen K, Humberset Ø, Bergström P, Agrup M, Dahl L, Gubanski M, Gojon H, Brahme CJ, Rydén I, Jakola AS, Vik-Mo EO, Lie HC, Asphaug L, Hervani M, Kristensen I, Rueegg CS, Olsen IC, Ledal RJ, Degsell E, Werlenius K, Blomstrand M, Brandal P (2023) Investigating survival, quality of life and cognition in PROton versus photon therapy for IDH-mutated diffuse grade 2 and 3 GLIOmas (PRO-GLIO): a randomised controlled trial in Norway and Sweden. BMJ Open 13:e070071. 10.1136/bmjopen-2022-070071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Heggebø LC, Borgen IMH, Blakstad H, Saxhaug C, Rønning PA, Niehusmann PF, Werlenius K, Blomstrand M, Brandal P (2025) Case report: Pseudoprogression mimicking neoplastic recurrence three months after completion of proton beam therapy for an IDH-mutant astrocytoma CNS WHO grade 3. Front Oncol 15:1397912. 10.3389/fonc.2025.1397912 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Hervey-Jumper SL, Zhang Y, Phillips JJ, Morshed RA, Young JS, McCoy L, Lafontaine M, Luks T, Ammanuel S, Kakaizada S, Egladyous A, Gogos A, Villanueva-Meyer J, Shai A, Warrier G, Rice T, Crane J, Wrensch M, Wiencke JK, Daras M, Oberheim Bush NA, Taylor JW, Butowski N, Clarke J, Chang S, Chang E, Aghi M, Theodosopoulos P, McDermott M, Jakola AS, Kavouridis VK, Nawabi N, Solheim O, Smith T, Berger MS, Molinaro AM (2023) Interactive effects of molecular, therapeutic, and patient factors on outcome of diffuse low-grade Glioma. J Clin Oncol 41:2029–2042. 10.1200/jco.21.02929 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ius T, Isola M, Budai R, Pauletto G, Tomasino B, Fadiga L, Skrap M (2012) Low-grade glioma surgery in eloquent areas: volumetric analysis of extent of resection and its impact on overall survival. A single-institution experience in 190 patients: clinical article. J Neurosurg 117:1039–1052. 10.3171/2012.8.Jns12393 [DOI] [PubMed] [Google Scholar]
- 27.Lauwens L, Ribeiro MF, Zegers CML, Høyer M, Alapetite C, Blomstrand M, Calugaru V, Perri DD, Iannalfi A, Lütgendorf-Caucig C, Paulsen F, Postma AA, Romero AM, Timmermann B, Troost EGC, van der Weide HL, Whitfield GA, Harrabi S, Lambrecht M, Eekers DBP (2025) Systematic review of MRI alterations in the brain following proton and photon radiation therapy: Towards a uniform European Particle Therapy Network (EPTN) definition. Radiother Oncol 208:110936. 10.1016/j.radonc.2025.110936 [DOI] [PubMed] [Google Scholar]
- 28.Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, Soffietti R, von Deimling A, Ellison DW (2021) The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol 23:1231–1251. 10.1093/neuonc/noab106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Lu VM, Welby JP, Laack NN, Mahajan A, Daniels DJ (2020) Pseudoprogression after radiation therapies for low grade glioma in children and adults: A systematic review and meta-analysis. Radiother Oncol 142:36–42. 10.1016/j.radonc.2019.07.013 [DOI] [PubMed] [Google Scholar]
- 30.Mellinghoff IK, Lu M, Wen PY, Taylor JW, Maher EA, Arrillaga-Romany I, Peters KB, Ellingson BM, Rosenblum MK, Chun S, Le K, Tassinari A, Choe S, Toubouti Y, Schoenfeld S, Pandya SS, Hassan I, Steelman L, Clarke JL, Cloughesy TF (2023) Vorasidenib and ivosidenib in IDH1-mutant low-grade Glioma: a randomized, perioperative phase 1 trial. Nat Med 29:615–622. 10.1038/s41591-022-02141-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mellinghoff IK, van den Bent MJ, Blumenthal DT, Touat M, Peters KB, Clarke J, Mendez J, Yust-Katz S, Welsh L, Mason WP, Ducray F, Umemura Y, Nabors B, Holdhoff M, Hottinger AF, Arakawa Y, Sepulveda JM, Wick W, Soffietti R, Perry JR, Giglio P, de la Fuente M, Maher EA, Schoenfeld S, Zhao D, Pandya SS, Steelman L, Hassan I, Wen PY, Cloughesy TF (2023) Vorasidenib in IDH1- or IDH2-mutant low-grade Glioma. N Engl J Med 389:589–601. 10.1056/NEJMoa2304194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mellinghoff IK, Penas-Prado M, Peters KB, Burris HA 3rd, Maher EA, Janku F, Cote GM, de la Fuente MI, Clarke JL, Ellingson BM, Chun S, Young RJ, Liu H, Choe S, Lu M, Le K, Hassan I, Steelman L, Pandya SS, Cloughesy TF, Wen PY (2021) Vorasidenib, a dual inhibitor of mutant IDH1/2, in recurrent or progressive Glioma; results of a first-in-human phase I trial. Clin Cancer Res 27:4491–4499. 10.1158/1078-0432.Ccr-21-0611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Merchant TE, Hua CH, Shukla H, Ying X, Nill S, Oelfke U (2008) Proton versus photon radiotherapy for common pediatric brain tumors: comparison of models of dose characteristics and their relationship to cognitive function. Pediatr Blood Cancer 51:110–117. 10.1002/pbc.21530 [DOI] [PubMed] [Google Scholar]
- 34.Meyers CA, Hess KR (2003) Multifaceted end points in brain tumor clinical trials: cognitive deterioration precedes MRI progression. Neuro Oncol 5:89–95. 10.1093/neuonc/5.2.89 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Miller JJ (2022) Targeting IDH-mutant Glioma. Neurotherapeutics 19:1724–1732. 10.1007/s13311-022-01238-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Millward CP, Armstrong TS, Barrington H, Brodbelt AR, Bulbeck H, Byrne A, Dirven L, Gamble C, Grundy PL, Islim AI, Javadpour M, Keshwara SM, Krishna ST, Mallucci CL, Marson AG, McDermott MW, Meling TR, Oliver K, Pizer B, Plaha P, Preusser M, Santarius T, Srikandarajah N, Taphoorn MJB, Watts C, Weller M, Williamson PR, Zadeh G, Zamanipoor Najafabadi AH, Jenkinson MD (2022) Opportunities and challenges for the development of “core outcome sets” in neuro-oncology. Neuro Oncol 24:1048–1055. 10.1093/neuonc/noac062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Montesinos P, Recher C, Vives S, Zarzycka E, Wang J, Bertani G, Heuser M, Calado RT, Schuh AC, Yeh SP, Daigle SR, Hui J, Pandya SS, Gianolio DA, de Botton S, Döhner H (2022) Ivosidenib and Azacitidine in IDH1-Mutated Acute Myeloid Leukemia. N Engl J Med 386:1519–1531. 10.1056/NEJMoa2117344 [DOI] [PubMed] [Google Scholar]
- 38.Muroi A, Mizumoto M, Ishikawa E, Ihara S, Fukushima H, Tsurubuchi T, Sakurai H, Matsumura A (2020) Proton therapy for newly diagnosed pediatric diffuse intrinsic pontine glioma. Childs Nerv Syst 36:507–512. 10.1007/s00381-019-04420-9 [DOI] [PubMed] [Google Scholar]
- 39.Natsume A, Arakawa Y, Narita Y, Sugiyama K, Hata N, Muragaki Y, Shinojima N, Kumabe T, Saito R, Motomura K, Mineharu Y, Miyakita Y, Yamasaki F, Matsushita Y, Ichimura K, Ito K, Tachibana M, Kakurai Y, Okamoto N, Asahi T, Nishijima S, Yamaguchi T, Tsubouchi H, Nakamura H, Nishikawa R (2023) The first-in-human phase I study of a brain-penetrant mutant IDH1 inhibitor DS-1001 in patients with recurrent or progressive IDH1-mutant Gliomas. Neuro Oncol 25:326–336. 10.1093/neuonc/noac155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Paganetti H, Zietman A (2015) Why Is Proton Beam Therapy So Controversial? J Am Coll Radiol 12:1318–1319. 10.1016/j.jacr.2015.09.019 [DOI] [PubMed] [Google Scholar]
- 41.Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, Chou R, Glanville J, Grimshaw JM, Hróbjartsson A, Lalu MM, Li T, Loder EW, Mayo-Wilson E, McDonald S, McGuinness LA, Stewart LA, Thomas J, Tricco AC, Welch VA, Whiting P, Moher D (2021) The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 372:n71. 10.1136/bmj.n71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL, Olivi A, McLendon R, Rasheed BA, Keir S, Nikolskaya T, Nikolsky Y, Busam DA, Tekleab H, Diaz LA Jr, Hartigan J, Smith DR, Strausberg RL, Marie SK, Shinjo SM, Yan H, Riggins GJ, Bigner DD, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW (2008) An integrated genomic analysis of human glioblastoma multiforme. Science 321:1807–1812. 10.1126/science.1164382 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Peters KB, Alford C, Heltemes A, Savelli A, Landi DB, Broadwater G, Desjardins A, Johnson MO, Low JT, Khasraw M, Ashley DM, Friedman HS, Patel MP (2024) Use, access, and initial outcomes of off-label ivosidenib in patients with IDH1 mutant Glioma. Neurooncol Pract 11:199–204. 10.1093/nop/npad068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Prasad V (2025) Surrogate end points in oncology: the speed-uncertainty trade-off from the patients’ perspective. Nat Rev Clin Oncol. 10.1038/s41571-025-01007-z [DOI] [PubMed] [Google Scholar]
- 45.Pulsifer MB, Sethi RV, Kuhlthau KA, MacDonald SM, Tarbell NJ, Yock TI (2015) Early Cognitive Outcomes Following Proton Radiation in Pediatric Patients With Brain and Central Nervous System Tumors. Int J Radiat Oncol Biol Phys 93:400–407. 10.1016/j.ijrobp.2015.06.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Preusser M, Geurts M, Canadian Cancer Trials Group, Cooperative Trials Group for Neuro-Oncology (COGNO) (2025) Vorasidenib maintenance for IDH mutant astrocytoma (VIGOR). ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT06809322. Accessed 30-03-2025
- 47.Qiu X, Gao J, Hu J, Yang J, Hu W, Huang Q, Zhang H, Lu JJ, Kong L (2023) Proton radiotherapy in the treatment of IDH-mutant diffuse gliomas: an early experience from shanghai proton and heavy ion center. J Neurooncol 162:503–514. 10.1007/s11060-022-04202-5 [DOI] [PubMed] [Google Scholar]
- 48.Retzer A, Baddeley E, Sivell S, Scott H, Nelson A, Bulbeck H, Seddon K, Grant R, Adams R, Watts C, Aiyegbusi OL, Kearns P, Rivera SC, Dirven L, Calvert M, Byrne A (2023) Development of a core outcome set for use in adult primary glioma phase III interventional trials: A mixed methods study. Neurooncol Adv 5:vdad096. 10.1093/noajnl/vdad096 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Retzer A, Sivell S, Scott H, Nelson A, Bulbeck H, Seddon K, Grant R, Adams R, Watts C, Aiyegbusi OL, Kearns P, Cruz Rivera S, Dirven L, Baddeley E, Calvert M, Byrne A (2022) Development of a core outcome set and identification of patient-reportable outcomes for primary brain tumour trials: protocol for the COBra study. BMJ Open 12:e057712. 10.1136/bmjopen-2021-057712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ritterbusch R, Halasz LM, Graber JJ (2021) Distinct imaging patterns of pseudoprogression in glioma patients following proton versus photon radiation therapy. J Neurooncol 152:583–590. 10.1007/s11060-021-03734-6 [DOI] [PubMed] [Google Scholar]
- 51.Rydén I, Carstam L, Gulati S, Smits A, Sunnerhagen KS, Hellström P, Henriksson R, Bartek J Jr, Salvesen Ø, Jakola AS (2020) Return to work following diagnosis of low-grade Glioma: a nationwide matched cohort study. Neurology 95:e856–e866. 10.1212/wnl.0000000000009982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Sagberg LM, Salvesen Ø, Jakola AS, Thurin E, De Dios E, Nawabi NLA, Kilgallon JL, Bernstock JD, Kavouridis VK, Smith TR, Solheim O (2024) Progression-free survival versus post-progression survival and overall survival in WHO grade 2 gliomas. Acta Oncol 63:798–804. 10.2340/1651-226x.2024.40845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sanai N, Chang S, Berger MS (2011) Low-grade Gliomas in adults. J Neurosurg 115:948–965. 10.3171/2011.7.Jns101238 [DOI] [PubMed] [Google Scholar]
- 54.Sankyo D (2020) A Study of DS-1001b in Patients With Chemotherapy- and Radiotherapy-Naive IDH1 Mutated WHO Grade II Glioma. ClinicalTrials.gov. https://clinicaltrials.gov/study/NCT04458272. Accessed 27 Feb 2025
- 55.Shih HA, Sherman JC, Nachtigall LB, Colvin MK, Fullerton BC, Daartz J, Winrich BK, Batchelor TT, Thornton LT, Mancuso SM, Saums MK, Oh KS, Curry WT, Loeffler JS, Yeap BY (2015) Proton therapy for low-grade gliomas: Results from a prospective trial. Cancer 121:1712–1719. 10.1002/cncr.29237 [DOI] [PubMed] [Google Scholar]
- 56.Slevin F, Hudson EM, Boele FW, Powell JR, Noutch S, Borland M, Brown S, Bruce A, Bulbeck H, Burnet NG, Chang YC, Colaco R, Currie S, Egleston D, Fersht N, Klein M, Lilley J, Lowe M, Miles E, Murray RD, O’Hara DJ, Norris M, Parbutt C, Smith A, Smith C, Whitfield GA, Short S, Murray L (2025) APPROACH: Analysis of Proton versus Photon Radiotherapy in Oligodendroglioma and Assessment of Cognitive Health - study protocol paper for a phase III multicentre, open-label randomised controlled trial. BMJ Open 15:e097810. 10.1136/bmjopen-2024-097810 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Tabrizi S, Yeap BY, Sherman JC, Nachtigall LB, Colvin MK, Dworkin M, Fullerton BC, Daartz J, Royce TJ, Oh KS, Batchelor TT, Curry WT, Loeffler JS, Shih HA (2019) Long-term outcomes and late adverse effects of a prospective study on proton radiotherapy for patients with low-grade glioma. Radiother Oncol 137:95–101. 10.1016/j.radonc.2019.04.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Taphoorn MJ (2003) Neurocognitive sequelae in the treatment of low-grade gliomas. Semin Oncol 30:45–48. 10.1053/j.seminoncol.2003.11.023 [DOI] [PubMed] [Google Scholar]
- 59.Teng KX, Price B, Joshi S, Alukaidey L, Shehab A, Mansour K, Toor GS, Angliss R, Drummond K (2021) Life after surgical resection of a low-grade glioma: A prospective cross-sectional study evaluating health-related quality of life. J Clin Neurosci 88:259–267. 10.1016/j.jocn.2021.03.038 [DOI] [PubMed] [Google Scholar]
- 60.Thurin E, Nyström PW, Smits A, Werlenius K, Bäck A, Liljegren A, Daxberg EL, Jakola AS (2018) Proton therapy for low-grade gliomas in adults: a systematic review. Clin Neurol Neurosurg 174:233–238. 10.1016/j.clineuro.2018.08.003 [DOI] [PubMed] [Google Scholar]
- 61.Tom MC, Cahill DP, Buckner JC, Dietrich J, Parsons MW, Yu JS (2019) Management for different Glioma subtypes: are all low-grade Gliomas created equal? Am Soc Clin Oncol Educ Book 39:133–145. 10.1200/edbk_238353 [DOI] [PubMed] [Google Scholar]
- 62.Tricco AC, Lillie E, Zarin W, O’Brien KK, Colquhoun H, Levac D, Moher D, Peters MDJ, Horsley T, Weeks L, Hempel S, Akl EA, Chang C, McGowan J, Stewart L, Hartling L, Aldcroft A, Wilson MG, Garritty C, Lewin S, Godfrey CM, Macdonald MT, Langlois EV, Soares-Weiser K, Moriarty J, Clifford T, Tunçalp Ö, Straus SE (2018) PRISMA extension for Scoping Reviews (PRISMA-ScR): checklist and explanation. Ann Intern Med 169:467–473. 10.7326/m18-0850 [DOI] [PubMed] [Google Scholar]
- 63.van den Bent MJ, Afra D, de Witte O, Ben Hassel M, Schraub S, Hoang-Xuan K, Malmström PO, Collette L, Piérart M, Mirimanoff R, Karim AB (2005) Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet 366:985–990. 10.1016/s0140-6736(05)67070-5 [DOI] [PubMed] [Google Scholar]
- 64.Walia A, Tuia J, Prasad V (2023) Progression-free survival, disease-free survival and other composite end points in oncology: improved reporting is needed. Nat Rev Clin Oncol 20:885–895. 10.1038/s41571-023-00823-5 [DOI] [PubMed] [Google Scholar]
- 65.Wick A, Bähr O, Schuler M, Rohrberg K, Chawla SP, Janku F, Schiff D, Heinemann V, Narita Y, Lenz HJ, Ikeda M, Ando Y, Wick W, Steinbach JP, Burger MC, Wenger K, Lassen U, Sankhala KK, Roggia C, Genvresse I, Munhoz C, Rentzsch C, Reschke S, Langer S, Wagner M, Kaulfuss S, Cai C, Lagkadinou E, Jeffers M, Peña C, Tabatabai G (2021) Phase I assessment of safety and therapeutic activity of BAY1436032 in patients with IDH1-mutant solid tumors. Clin Cancer Res 27:2723–2733. 10.1158/1078-0432.Ccr-20-4256 [DOI] [PubMed] [Google Scholar]
- 66.Winter SF, Gardner MM, Karschnia P, Vaios EJ, Grassberger C, Bussière MR, Nikolic K, Pongpitakmetha T, Ehret F, Kaul D, Boehmerle W, Endres M, Shih HA, Parsons MW, Dietrich J (2024) Unique brain injury patterns after proton vs photon radiotherapy for WHO grade 2–3 gliomas. Oncologist 29:e1748–e1761. 10.1093/oncolo/oyae195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Yavas C, Zorlu F, Ozyigit G, Gurkaynak M, Yavas G, Yuce D, Cengiz M, Yildiz F, Akyol F (2012) Prospective assessment of health-related quality of life in patients with low-grade glioma: a single-center experience. Support Care Cancer 20:1859–1868. 10.1007/s00520-011-1288-4 [DOI] [PubMed] [Google Scholar]
- 68.Yu H, He S, He Y, Dai G, Fu Y, Zeng X, Liu M, Ai P (2024) Dosimetric comparison of advanced radiation techniques for scalp-sparing in low-grade gliomas. Strahlenther Onkol 200:785–796. 10.1007/s00066-024-02229-3 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
(PDF 97.0 KB)
Data Availability Statement
No datasets were generated or analysed during the current study.

