Comparison of Clazosentan and Nimodipine on Vasospasm and Vasospasm-Related Outcomes after Aneurysmal Subarachnoid Hemorrhage : A Post-hoc Propensity Score-Matched Analysis of Six Randomized Clinical Trials

Article information

J Korean Neurosurg Soc. 2025;.jkns.2024.0195
Publication date (electronic) : 2025 January 17
doi : https://doi.org/10.3340/jkns.2024.0195
1Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
2Department of Neurosurgery, Hanyang University Medical Center, Hanyang University College of Medicine, Seoul, Korea
3Data Management & Biometry∙Statistics Division, Nxera Pharma Japan Co., Ltd., Tokyo, Japan
4Department of Neurosurgery, Bucheon St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
Address for reprints : Ik Seong Park Department of Neurosurgery, Bucheon St. Mary’s Hospital, The Catholic University of Korea, 327 Sosa-ro, Bucheon 14647, Korea Tel : +82-32-340-7212, Fax : +82-32-340-7391, E-mail : ispahk@gmail.com
*Sung Ho Lee and Kyu-Sun Choi contributed equally as first authors.
Received 2024 October 26; Accepted 2024 December 17.

Abstract

Objective

Clazosentan is a recently approved endothelin receptor antagonist indicated for the prevention of vasospasm and related complications following aneurysmal subarachnoid hemorrhage (aSAH). To date, no direct, head-to-head comparison between clazosentan and nimodipine has been conducted. In this study, we indirectly assessed the efficacy and safety of these two drugs in preventing vasospasm and its associated outcomes after aSAH.

Methods

Participants from six randomized clinical trials of clazosentan were reclassified into three subgroups based on their concomitant use of oral nimodipine : 1) a clazosentan subgroup (without nimodipine), 2) a nimodipine subgroup (without clazosentan), and 3) a placebo subgroup (receiving neither clazosentan nor nimodipine). Data from participants who received the approved dose of clazosentan 10 mg/h was analyzed. To account for heterogeneities among the analyzed studies, we performed within-study comparisons of subgroups and pooled data from the same subgroup. To further balance the three groups, we conducted a propensity score-matching and compared the outcomes among subgroups. The outcomes measured were angiographic vasospasm within 14 days after aSAH and vasospasm-related morbidity and all-cause mortality (MM) within 6 weeks, defined as death, vasospasm-related new cerebral infarcts, delayed ischemic neurological deficits, or initiation of rescue therapy. Incidence and relative risk reduction (RRR) were analyzed across subgroups, and overall safety was reviewed.

Results

The pooled data from within-study comparisons demonstrated that clazosentan significantly reduced the risk of vasospasm (RRR, 0.48; 95% confidence interval [CI], 0.35 to 0.58) and MM (RRR, 0.47; 95% CI, 0.30 to 0.60) compared to placebo, whereas nimodipine did not. In the propensity score-matched analysis, clazosentan demonstrated a significant risk reduction in outcomes when compared to nimodipine (RRR, 0.63; 95% CI, 0.46 to 0.75 for vasospasm; RRR, 0.29; 95% CI, 0.04 to 0.48 for MM) and placebo (RRR, 0.59; 95% CI, 0.40 to 0.72 for vasospasm; RRR, 0.41; 95% CI, 0.21 to 0.56 for MM).The overall safety results were comparable across the three subgroups and consistent with the expected range for endothelin receptor antagonists.

Conclusion

Clazosentan at 10 mg/h significantly reduced the incidence of cerebral vasospasm and MM following aSAH, compared to both placebo and nimodipine. Further clinical studies are warranted to compare the efficacy of clazosentan and nimodipine to optimize treatment strategies for aSAH.

INTRODUCTION

Aneurysmal subarachnoid hemorrhage (aSAH) is a critical neurological emergency caused by the rupture of an intracranial aneurysm. Despite advances in early surgical interventions such as surgical clipping and endovascular coiling [20], cerebral vasospasm occurs in up to 70% of aSAH patients, significantly increasing the risk of delayed cerebral ischemia (DCI), which exacerbates neurological injury [31].

For the pharmacological prevention of DCI following aSAH, various international organizations such as the American Heart Association/American Stroke Association (AHA/ASA), European Stroke Organization, and Korean Society of Cerebrovascular Surgeons recommend the oral administration of nimodipine [6,15,33]. However, in Japan, nimodipine is neither approved nor recommended as standard care, according to the Japan Stroke Society guidelines [17].

Although nimodipine is widely used, there is a lack of data comparing the incidence of vasospasm and related complications before and after its introduction. Additionally, the prognosis of aSAH remains poor, with cerebral vasospasm and DCI posing persistent challenges. Even during the widespread use of nimodipine, studies report that approximately 20–50% of patients still develop DCI after aSAH [7], and cerebral vasospasm accounts for 33.7% of deaths within 90 days following aSAH [18]. In patients unresponsive to pharmacological treatments, endovascular therapy is currently the only available option. These limited treatment options underscore the urgent need for new therapeutic strategies to more effectively address vasospasm and DCI.

Clazosentan (PIVLAZ®), a highly selective endothelin receptor antagonist, was approved in Japan (January 2022) [26] and in South Korea (December 2023) [27] for the prevention of cerebral vasospasm and vasospasm-related cerebral infarction or cerebral ischemic episodes after aSAH. Unlike nimodipine, clazosentan specifically targets endothelin A (ETA) receptors, counteracting the potent vasoconstrictive effects of endothelin-1 (ET-1), which plays a key role in the pathophysiology of cerebral vasospasm following aSAH [19].

Currently, no direct head-to-head comparison between clazosentan and nimodipine following aSAH has been reported. A direct, head-to-head comparison between nimodipine and clazosentan may not be feasible given current clinical practices. Therefore, we performed an exploratory post-hoc analysis using data from six randomized controlled trials (RCTs) of clazosentan to indirectly compare its efficacy with that of nimodipine in preventing vasospasm and vasospasm-related morbidity and all-cause mortality (MM).

MATERIALS AND METHODS

Each randomized controlled trial included in our analyses was approved by the appropriate Institutional Review Board or Ethics Committee and conducted in accordance with the Declaration of Helsinki and Good Clinical Practice guidelines. Our secondary analyses were conducted using anonymized data and did not require additional ethical approval.

Data sources

A post-hoc analysis was conducted using data from six RCTs of clazosentan with similar designs : two phase 2 studies (AC-054-201 [CONSCIOUS-1 [22]] and AC-054-202 [11] [hereafter 201 and 202]) and four phase 3 studies (AC-054-301 [CONSCIOUS-2 [19]], AC-054-302 [CONSCIOUS-3 [20]], AC-054-305, and AC-054-306 [10] [hereafter 301, 302, 305, and 306]). Comprehensive details on study designs and eligibility criteria have been published elsewhere [10,11,19-22] and are summarized in Table 1.

Summary of analyzed studies

In these six studies, participants were administered clazosentan intravenously at doses of 1, 5, 10, or 15 mg/h, or a placebo, within 48 to 56 hours of aSAH onset. Treatment continued for up to 14 days following the onset of aSAH. Orally administered nimodipine was permitted as background therapy, except for participants enrolled in Japan. Intra-arterial vasodilators, including nimodipine, were classified as rescue medications, and the prophylactic administration of intravenous nimodipine was prohibited by study protocols.

Study population and reclassification of subgroups

For this post-hoc analysis, participants from six clinical studies were reclassified into three subgroups based on their concomitant use of oral nimodipine : 1) a clazosentan subgroup consisting of those who received clazosentan without nimodipine, 2) a nimodipine subgroup, consisting of those who received placebo along with oral nimodipine, and 3) a placebo subgroup, consisting of those who received placebo without nimodipine (Fig. 1). In the clazosentan subgroup, data from participants who received the 10 mg/h dose of clazosentan, the approved dose in Korean and Japan, were used for efficacy analysis. For overall safety analysis, data from participant who received clazosentan were used, regardless of dose. In the nimodipine subgroup, participants were included if they received nimodipine from 24 hours before the initiation of study treatment and up to 24 hours after its discontinuation. Participants who received nimodipine as a rescue medication were excluded from the nimodipine subgroup.

Fig. 1.

Reclassification of six randomized clinical trials of clazosentan. *Among participants who were originally allocated in the clazosentan arm, those who received nimodipine concomitantly were not included in this post-hoc analysis. Participants who received the approved dose of 10 mg/h in Korean and Japan were included in this post-hoc analysis. In the nimodipine subgroup, participants were included if they received nimodipine 24 hours before the initiation of study treatment and up to 24 hours after its discontinuation. Participants who received nimodipine as a rescue medication were excluded from the nimodipine subgroup. RCT : randomized controlled trial.

For the efficacy analysis evaluating vasospasm, participants from Studies 201, 202, 305, and 306 were used, as angiographic vasospasm was assessed in these four studies. However, for the analysis of MM and the safety analysis, participants from all of six studies were included.

Outcomes

The analysis focused on two key outcomes : the difference in the incidence of moderate to severe angiographic vasospasm within 14 days after aSAH between subgroups and the difference in the incidence of MM within 6 weeks after aSAH between subgroups. Moderate to severe angiographic vasospasm was defined as a reduction of ≥34% in the inner artery diameter, as assessed by digital subtraction angiography images and determined by Image Review Committees. The MM were defined as vasospasm-related new cerebral infarcts, delayed ischemic neurological deficits (DINDs), rescue therapy initiation, and death from any cause within 6 weeks following aSAH. DINDs were defined as a decline of 2 or more points on the modified Glasgow coma scale or a rise of 2 or more points on the abbreviated National Institutes of Health stroke scale that lasted at least 2 hours. Rescue therapy was initiated when clinical or imaging signs indicated vasospasm. In Studies 305 and 306, the initiation of rescue therapy was not initially considered as MM. However, in this post-hoc analysis, we have included the initiation of rescue therapy as a component of MM. All MM were centrally reviewed by expert committees, such as the Critical Events Committee or Events Review Committee, within each study to ensure consistent assessment across the respective study sites [11,12,21,22,24,34]. Additionally, safety outcomes were evaluated, including the incidence of treatment-emergent adverse events (TEAEs), serious TEAEs, TEAEs leading to treatment discontinuation, and main causes of death. TEAEs were collected up to 24 hours after study treatment discontinuation in all studies. Data was collected up to 12 weeks post-aSAH for death. TEAEs were coded using Medical Dictionary for Regulatory Activities (MedDRA) version 22.1.

Within-study comparison and subgroup pooled data

Since the baseline characteristics and clinical outcomes differed among the six RCTs (as shown in Table 1), comparisons between subgroups were first conducted separately within each trial, and results were presented by individual studies. After completing within-study comparisons, data from the same subgroups were pooled across studies to calculate an overall event rate for each subgroup. The pooled data were then used to calculate the relative risk reduction (RRR) between subgroups, comparing clazosentan vs. placebo and nimodipine vs. placebo. Since Study 202 was the only study in which both clazosentan 10 mg and nimodipine were administered, a comparison between the clasozentan and nimodipine subgroups was conducted using propensity-score matched analysis.

Propensity score-matched analysis

Given that participants were not randomly assigned to receive nimodipine, propensity score matching was performed using greedy nearest neighbor matching to balance baseline characteristics across participants. The propensity score was calculated using the logistic regression model, and matching was performed using calipers of width equal to 0.2 times the standard deviation of the logit of the propensity scores with an equal ratio between clazosentan and nimodipine subgroups, followed by clazosentan and placebo subgroups. First, participants who received clazosentan at a dose of 10 mg/h were matched with those in the nimodipine subgroup. Next, the matched participants in the matched clazosentan subgroup were further matched with those in the placebo subgroup. The propensity score was calculated using covariates selected based on their p-values of less than 0.1 in univariate analyses (World Federation of Neurosurgical Societies [WFNS] grade, clot size, age, and body mass index), or based on clinical significance in aSAH (securing procedures). Analyses comparing clazosentan vs. placebo, nimodipine vs. placebo, and clazosentan vs. nimodipine were conducted using Fisher’s exact test.

Statistical analysis

All statistical analyses were performed using SAS® release 9.4 for Windows (SAS Institute Inc., Cary, NC, USA). The RRR of outcomes was provided with corresponding 95% confidence intervals (CIs). Continuous variables were compared using a two-paired t-test, and proportions were compared using Fisher’s exact test, unless otherwise specified.

RESULTS

Baseline characteristics of analyzed studies

Six randomized studies included a total of 2746 participants aged 18–75 years with aSAH, who underwent either surgical clipping or endovascular coiling (Table 1). Among the participants, 277 received 10 mg/h clazosentan, while 949 received a placebo. The use of nimodipine varied across studies : it was not used in Studies 305 and 306 (conducted in Japan), was used in 19% of participants in Study 202 (conducted in Korea and Japan), and in 80–95% of participants in the other studies. The eligibility criteria for Studies 202, 305, and 306 specified Fisher grade 3, while the other studies specified WFNS grade I-IV. However, baseline clot sizes were generally consistent with Fisher grade 3 or involved a blood clot of equivalent size (short axis ≥1 mm) in all studies, aligning with the labeled indication for clazosentan. Angiographic vasospasm was evaluated in Studies 201, 202, 305, and 306, while MM were reported in all six studies.

Within-study comparison and subgroup pooled data

Analysis for vasospasm included data from Studies 202, 305, and 306 for the clazosentan 10 mg/h subgroup, and Studies 201 and 202 for the nimodipine subgroup. In the clazosentan subgroup, vasospasm occurred in 29.59% (79/267) of participants, compared to 56.55% (151/267) in the placebo subgroup (Table 2). This demonstrated a statistically significant RRR of 0.48 (95% CI, 0.35 to 0.58). In the nimodipine subgroup, vasospasm occurred in 68.04% (66/97) of participants compared to 79.31% (46/58) in the placebo subgroup, yielding an RRR of 0.14 (95% CI, -0.04 to 0.29), which was not statistically significant.

Within-study comparison and subgroup pooled results of vasospasm and vasospasm-related morbidity and mortality

Analysis for MM included data from Studies 202, 305, and 306 for the clazosentan 10 mg/h subgroup, and Studies 201, 202, 301, and 302 for the nimodipine subgroup. In the clazosentan subgroup, MM occurred in 20.60% (55/267) of participants, compared to 38.95% (104/267) in the placebo subgroup (Table 2). This demonstrated a statistically significant RRR of 0.47 (95% CI, 0.30 to 0.60). In the nimodipine subgroup, MM occurred in 28.19% (170/603) of participants compared to 35.48% (44/124) in the placebo group, yielding an RRR of 0.21 (95% CI, -0.04 to 0.39), which was not statistically significant.

Propensity score-matched analysis

The propensity score distributions for matched groups were acceptable between the clazosentan and nimodipine or placebo subgroups (Fig. 2). The matched analysis for vasospasm and MM included 89 and 231 participants in each group, respectively. The baseline characteristics of the matched subgroups are shown in Table 3.

Fig. 2.

Propensity score distributions. Propensity score density histogram of each subgroup used to analyze (A) moderate to severe angiographic vasospasm within 14 days after aSAH and (B) vasospasm-related morbidity and all-cause mortality within 6 weeks after aSAH. aSAH : aneurysmal subarachnoid hemorrhage.

Baseline characteristics of participants in the propensity score-matched analysis

Vasospasm occurred in 25.84% (23/89) of participants in the matched clazosentan subgroup, compared to 69.66% (62/89) in the matched nimodipine subgroup and 62.92% (56/89) in the matched placebo subgroup (Fig. 3A). The RRR was statistically significant between clazosentan and placebo (0.59; 95% CI, 0.40 to 0.72) and clazosentan and nimodipine (0.63; 95% CI, 0.46 to 0.75).

Fig. 3.

Propensity score-matched analysis for angiographic vasospasm and vasospasm-related morbidity and all-cause mortality. Event rate (%) and relative risk reductions (95% CIs) are shown for (A) moderate to severe angiographic vasospasm within 14 days after aSAH and (B) vasospasm-related morbidity and all-cause mortality within 6 weeks after aSAH. The numbers on each bar represent the event rates (%) for each subgroup. Only statistically significant p-values based on Fisher’s exact test between two groups are displayed : otherwise, not significant. CI : confidence interval, aSAH : aneurysmal subarachnoid hemorrhage.

MM occurred in 22.08% (51/231) of participants in the matched clazosentan subgroup, compared to 31.17% (72/231) in the matched nimodipine subgroup and 37.66% (87/231) in the matched placebo subgroup (Fig. 3B). The RRR was statistically significant between clazosentan and placebo (0.41; 95% CI, 0.21 to 0.56) and clazosentan and nimodipine (0.29; 95% CI, 0.04 to 0.48).

Overall safety

In the pooled safety analysis, the incidences of TEAEs were comparable among the clazosentan (93.1%), nimodipine (91.3%), and placebo (93.9%) subgroups, while the incidences of serious TEAEs were higher in nimodipine subgroup (34.7%) than clazosentan (23.8%) and placebo (19.6%) subgroups. Deaths were reported in 5.0% (24/479), 4.6% (28/606) and 3.7% (13/347) of clazosentan, nimodipine and placebo subgroups, respectively. Main causes of death included cerebral infarction, cerebral ischemia, cerebral vasoconstriction and brain edema (Table 4).

Safety overview

Among TEAEs, anemia, hypokalemia, pleural effusion, pulmonary edema and cerebral ischemia were more common in clazosentan subgroup compared to nimodipine and placebo subgroups. In contrast, pneumonia, urinary tract infection, hepatic enzyme increased, and hydrocephalus were reported more frequently in nimodipine subgroup (Table 5).

TEAEs reported by ≥3% of clazosentan-treated patients

DISCUSSION

Since global guidelines, including those from the USA, EU, and Korea, recommend nimodipine as the first-line treatment choice after aSAH [6,15,33], supported by a high level of evidence, it is already widely and routinely used in clinical practice. Given this context, conducting a direct head-to-head comparison with the newly approved clazosentan, which has a different mechanism of action, is challenging. At present, a post-hoc analysis using the existing six RCTs offers the most feasible approach.

Therefore, we indirectly assessed the efficacy and safety of these two drugs in preventing vasospasm and its associated outcomes after aSAH using the post-hoc analysis. Within-study comparisons and subgroup pooled data showed that clazosentan 10 mg/h significantly reduced the risk of vasospasm and MM compared to placebo, whereas nimodipine did not achieve statistical significance. Furthermore, in the propensity score-matched analysis, clazosentan 10 mg/h demonstrated a significant risk reduction in cerebral vasospasm and related outcomes when compared to both nimodipine and placebo. However, nimodipine did not yield a statistically significant benefit over placebo. These results suggest that clazosentan could provide a new strategy for preventing vasospasm and related complications. Further comparisons with nimodipine are needed to fully understand their relative efficacy. Throughout its clinical development program, clazosentan has demonstrated angiographic evidence of reducing vasospasm in several key trials (Studies 201, 202, 305, and 306). Furthermore, a recent meta-analysis by Pontes et al. [31] reported that clazosentan significantly reduced vasospasm-related DCI (relative risk [RR], 0.56; 95% CI, 0.38 to 0.81) and angiographic vasospasm (RR, 0.54; 95% CI, 0.47 to 0.61) compared to placebo across seven RCTs, six of which were included in our analysis.

The mechanism of action and evidence base for clazosentan and nimodipine differs in several important aspects. First, nimodipine, an L-type calcium channel antagonist, inhibits the influx of calcium ions in vascular smooth muscles, thereby promoting vasodilation. However, the mechanism of action of nimodipine on the prevention of DCI in aSAH is uncertain [5]. In contrast, clazosentan has a distinct mechanism of action by antagonizing ET-1 and preventing vasoconstriction that contributes to cerebral vasospasm [7]. Second, oral nimodipine lacks sufficient and up-to-date evidence [9,13]. The beneficial effect of oral nimodipine was first reported in 1983 [1] and subsequently investigated in several studies during the 1980s [24,25,28-30], which provided the foundation for its recommendation in global guidelines [6,15,33]. Conversely, clazosentan’s efficacy has been confirmed more recently in patients who underwent endovascular coiling (Study 305) and surgical clipping (Study 306) for aSAH, with data collected between 2016 and 2020. Third, while the clinical data for oral nimodipine primarily originate from Western populations (USA, Cananda, France, and the UK), clazosentan’s development program included a substantial number of patients from Japan (440 patients enrolled in Studies 305 and 306, 105 patients in Study 202) and South Korea (74 patients in Study 202). This broader and more recent demographic representation provides a more comprehensive assessment of clazosentan’s efficacy across diverse populations.

In this analysis, the RRR for vasospasm and MM with nimodipine was not statistically significant compared to placebo, which may be partially attributed to uncontrolled drug administration. Retrospective studies [8,14,32] have suggested that dose reduction or interruption of oral nimodipine is associated with unfavorable outcomes, such as an increased incidence of DCI. However, in the six RCTs analyzed, nimodipine was administered according to best clinical practice, guided by physician discretion and local regulations. Unfortunately, detailed data on dosage and treatment duration were limited. The relatively small number of subjects, particularly in the pooled analysis for vasospasm (58 participants in the placebo group), may have contributed to the marginal effect observed with nimodipine. Similarly, the beneficial effect of oral nimodipine is largely based on a single major study [30] with approximately 270 participants in each group. When this largest study is excluded, meta-analyses fail to demonstrate statistical significance, with the remaining studies including only 30–80 subjects per group [24,25,28-30].

Although the RCTs in our analysis primarily enrolled patients with Fisher grade 3 hemorrhage, the baseline characteristics, particularly clot size and outcomes, varied across the six studies. For example, over 90% of patients in Study 202 had diffuse thick clots, compared to approximately 50% in Studies 305 and 306, which may have contributed to the better outcome observed in the latter studies. Additionally, in the pooled analysis for vasospasm, approximately 80% of patients in the placebo groups of Studies 201 and 202 experienced vasospasms, compared to 50% in Studies 305 and 306. Given the heterogeneity in baseline characteristics and varying event rates, comparisons were made within subgroups of individual studies rather than pooling the results from all studies.

In the pooled safety analysis, the overall incidences of TEAEs were comparable among the subgroups. Clazosentan was associated with a higher incidence of pleural effusion (12.7%) and pulmonary edema (8.1%) compared to placebo (3.5% and 4.3%, respectively) and nimodipine (4.3% and 4.5%, respectively). However, most patients who developed pleural effusion or pulmonary edema were adequately managed and recovered during hospitalization, with no fatal cases reported. These adverse events are consistent with the known class effects of ETA receptor antagonists, with similar or lower placebo-corrected incidence rates of edema or fluid overload observed with other agents such as zibotentan (38%), atrasentan (28%), ambrisentan (18%), avosentan (15%), durasentan (12%), and sitaxentan (8%) [2-4,12,16,23,35].

Our study has several strengths, including the indirect comparison of clazosentan, a newly approved ETA receptor antagonist, with nimodipine. To address the heterogeneity of enrolled subjects and clinical outcomes, we conducted a propensity score-matched analysis to ensure more balanced treatment groups and minimize bias. In the propensity score-matched analysis, 89 and 231 participants were selected from each subgroup for vasospasm and MM, respectively, which are relatively large sample sizes compared to previous studies investigating nimodipine.

However, our study also has limitations including the uncontrolled administration of oral nimodipine and the lack of detailed data on its usage. Furthermore, heterogeneity in the eligibility criteria, baseline characteristics and changes in the standard of care, such as the removal of hypervolemia as a recommended treatment since 2012 [15], may have influenced our results. These limitations highlight the need for further well-designed clinical studies to compare the efficacy and safety of clazosentan and nimodipine, given the evolving standards of care in aSAH management.

CONCLUSION

In this exploratory post-hoc analysis of six RCTs, clazosentan 10 mg/h demonstrated a significant reduction in the incidence of cerebral vasospasm and MM following aSAH, compared to both placebo and nimodipine. The overall safety results were comparable across the three subgroups and consistent with the expected range for ETA receptor antagonists. The TEAEs of pleural effusion and pulmonary edema were generally well managed, despite their higher incidences with clazosentan. While nimodipine has long been used, our findings suggest that clazosentan may offer superior efficacy. Further clinical studies comparing the efficacy of clazosentan, either alone or in combination with nimodipine, are needed to determine the potential treatment position of clazosentan and to develop optimal treatment strategies for aSAH.

Notes

Conflicts of interest

No potential conflict of interest relevant to this article was reported.

Informed consent

This type of study does not require informed consent.

Author contributions

Conceptualization : ISP; Data curation : KSC, SHL, ISP; Formal analysis : OT; Funding acquisition : ISP; Methodology : KSC, SHL, OT; Project administration : OT, ISP; Visualization : OT; Writing - original draft : KSC, SHL, OT; Writing - review & editing : OT, ISP

Data sharing

None

Preprint

None

Acknowledgements

The authors acknowledge AIMS BioScience (www.aimsbiosci.com) for providing medical writing support.

References

1. Allen GS, Ahn HS, Preziosi TJ, Battye R, Boone SC, Boone SC, et al. Cerebral arterial spasm--a controlled trial of nimodipine in patients with subarachnoid hemorrhage. N Engl J Med 308:619–624. 1983;
2. Barst RJ, Langleben D, Frost A, Horn EM, Oudiz R, Shapiro S, et al. Sitaxsentan therapy for pulmonary arterial hypertension. Am J Respir Crit Care Med 169:441–447. 2004;
3. Black HR, Bakris GL, Weber MA, Weiss R, Shahawy ME, Marple R, et al. Efficacy and safety of darusentan in patients with resistant hypertension: results from a randomized, double-blind, placebo-controlled dose-ranging study. J Clin Hypertens (Greenwich) 9:760–769. 2007;
4. Carducci MA, Saad F, Abrahamsson PA, Dearnaley DP, Schulman CC, North SA, et al. A phase 3 randomized controlled trial of the efficacy and safety of atrasentan in men with metastatic hormone-refractory prostate cancer. Cancer 110:1959–1966. 2007;
5. Caylor MM, Macdonald RL. Pharmacological prevention of delayed cerebral ischemia in aneurysmal subarachnoid hemorrhage. Neurocrit Care 40:159–169. 2024;
6. Cho WS, Kim JE, Park SQ, Ko JK, Kim DW, Park JC, et al. Korean clinical practice guidelines for aneurysmal subarachnoid hemorrhage. J Korean Neurosurg Soc 61:127–166. 2018;
7. Daou BJ, Koduri S, Thompson BG, Chaudhary N, Pandey AS. Clinical and experimental aspects of aneurysmal subarachnoid hemorrhage. CNS Neurosci Ther 25:1096–1112. 2019;
8. Ditz C, Matone MV, Schwachenwald B, Küchler J. Risks of nimodipine dose reduction during the high-risk period for delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. Neurosurg Rev 47:37. 2024;
9. Dorhout Mees SM, Rinkel GJ, Feigin VL, Algra A, van den Bergh WM, Vermeulen M, et al. Calcium antagonists for aneurysmal subarachnoid haemorrhage. Cochrane Database Syst Rev 2007:CD000277. 2007;
10. Endo H, Hagihara Y, Kimura N, Takizawa K, Niizuma K, Togo O, et al. Effects of clazosentan on cerebral vasospasm-related morbidity and all-cause mortality after aneurysmal subarachnoid hemorrhage: two randomized phase 3 trials in Japanese patients. J Neurosurg 137:1707–1717. 2022;
11. Fujimura M, Joo JY, Kim JS, Hatta M, Yokoyama Y, Tominaga T. Preventive effect of clazosentan against cerebral vasospasm after clipping surgery for aneurysmal subarachnoid hemorrhage in Japanese and Korean patients. Cerebrovasc Dis 44:59–67. 2017;
12. Galiè N, Olschewski H, Oudiz RJ, Torres F, Frost A, Ghofrani HA, et al. Ambrisentan for the treatment of pulmonary arterial hypertension: results of the ambrisentan in pulmonary arterial hypertension, randomized, double-blind, placebo-controlled, multicenter, efficacy (ARIES) study 1 and 2. Circulation 117:3010–3019. 2008;
13. Hao G, Chu G, Pan P, Han Y, Ai Y, Shi Z, et al. Clinical effectiveness of nimodipine for the prevention of poor outcome after aneurysmal subarachnoid hemorrhage: a systematic review and meta-analysis. Front Neurol 13:982498. 2022;
14. Hernández-Durán S, Mielke D, Rohde V, Malinova V. Does nimodipine interruption due to high catecholamine doses lead to a greater incidence of delayed cerebral ischemia in the setting of aneurysmal subarachnoid hemorrhage? World Neurosurg 132:e834–e840. 2019;
15. Hoh BL, Ko NU, Amin-Hanjani S, Chou SH-Y, Cruz-Flores S, Dangayach NS, et al. 2023 guideline for the management of patients with aneurysmal subarachnoid hemorrhage: a guideline from the American Heart Association/ American Stroke Association. Stroke 54:e314–e370. 2023;
16. James ND, Caty A, Borre M, Zonnenberg BA, Beuzeboc P, Morris T, et al. Safety and efficacy of the specific endothelin-A receptor antagonist ZD4054 in patients with hormone-resistant prostate cancer and bone metastases who were pain free or mildly symptomatic: a double-blind, placebo-controlled, randomised, phase 2 trial. Eur Urol 55:1112–1123. 2009;
17. Japan Stroke Society : Japan Stroke Society Guideline 2021 for the Treatment of Stroke. Available at : https://www.jsts.gr.jp/img/guideline2021_kaitei2023.pdf.
18. Khanafer A, Bhogal P, Hellstern V, Harmening C, Bäzner H, Ganslandt O, et al. Vasospasm-related death after aneurysmal subarachnoid hemorrhage: a retrospective case-control study. J Clin Med 11:4642. 2022;
19. Macdonald RL, Higashida RT, Keller E, Mayer SA, Molyneux A, Raabe A, et al. Clazosentan, an endothelin receptor antagonist, in patients with aneurysmal subarachnoid haemorrhage undergoing surgical clipping: a randomised, double-blind, placebo-controlled phase 3 trial (CONSCIOUS-2). Lancet Neurol 10:618–625. 2011;
20. Macdonald RL, Higashida RT, Keller E, Mayer SA, Molyneux A, Raabe A, et al. Randomized trial of clazosentan in patients with aneurysmal subarachnoid hemorrhage undergoing endovascular coiling. Stroke 43:1463–1469. 2012;
21. Macdonald RL, Higashida RT, Keller E, Mayer SA, Molyneux A, Raabe A, et al. Preventing vasospasm improves outcome after aneurysmal subarachnoid hemorrhage: rationale and design of CONSCIOUS-2 and CONSCIOUS-3 trials. Neurocrit Care 13:416–424. 2010;
22. Macdonald RL, Kassell NF, Mayer S, Ruefenacht D, Schmiedek P, Weidauer S, et al. Clazosentan to overcome neurological ischemia and infarction occurring after subarachnoid hemorrhage (CONSCIOUS-1): randomized, double-blind, placebo-controlled phase 2 dose-finding trial. Stroke 39:3015–3021. 2008;
23. Mann JF, Green D, Jamerson K, Ruilope LM, Kuranoff SJ, Littke T, et al. Avosentan for overt diabetic nephropathy. J Am Soc Nephrol 21:527–535. 2010;
24. Mee E, Dorrance D, Lowe D, Neil-Dwyer G. Controlled study of nimodipine in aneurysm patients treated early after subarachnoid hemorrhage. Neurosurgery 22:484–491. 1988;
25. Neil-Dwyer G, Mee E, Dorrance D, Lowe D. Early intervention with nimodipine in subarachnoid haemorrhage. Eur Heart J 8 Suppl K:41–47. 1987;
26. Nxera Pharma Japan Co., Ltd. : PIVLAZ®. Package Insert. Tokyo : Nxera Pharma Japan Co., Ltd., 2022.
27. Nxera Pharma Korea Co., Ltd. : PIVLAZ®. Package Insert. Seoul : Nxera Pharma Japan Co., Ltd., 2023.
28. Petruk KC, West M, Mohr G, Weir BK, Benoit BG, Gentili F, et al. Nimodipine treatment in poor-grade aneurysm patients. Results of a multicenter double-blind placebo-controlled trial. J Neurosurg 68:505–517. 1988;
29. Philippon J, Grob R, Dagreou F, Guggiari M, Rivierez M, Viars P. Prevention of vasospasm in subarachnoid haemorrhage. A controlled study with nimodipine. Acta Neurochir (Wien) 82:110–114. 1986;
30. Pickard JD, Murray GD, Illingworth R, Shaw MD, Teasdale GM, Foy PM, et al. Effect of oral nimodipine on cerebral infarction and outcome after subarachnoid haemorrhage: British aneurysm nimodipine trial. BMJ 298:636–642. 1989;
31. Pontes JPM, Santos MDC, Gibram FC, Rodrigues NMV, Cavalcante-Neto JF, Barros ADM, et al. Efficacy and safety of clazosentan after aneurysmal subarachnoid hemorrhage: an updated meta-analysis. Neurosurgery 93:1208–1219. 2023;
32. Sandow N, Diesing D, Sarrafzadeh A, Vajkoczy P, Wolf S. Nimodipine dose reductions in the treatment of patients with aneurysmal subarachnoid hemorrhage. Neurocrit Care 25:29–39. 2016;
33. Steiner T, Juvela S, Unterberg A, Jung C, Forsting M, Rinkel G, et al. European Stroke Organization guidelines for the management of intracranial aneurysms and subarachnoid haemorrhage. Cerebrovasc Dis 35:93–112. 2013;
34. Vajkoczy P, Meyer B, Weidauer S, Raabe A, Thome C, Ringel F, et al. Clazosentan (AXV-034343), a selective endothelin A receptor antagonist, in the prevention of cerebral vasospasm following severe aneurysmal subarachnoid hemorrhage: results of a randomized, double-blind, placebo-controlled, multicenter phase IIa study. J Neurosurg 103:9–17. 2005;
35. Vercauteren M, Trensz F, Pasquali A, Cattaneo C, Strasser DS, Hess P, et al. Endothelin ETA receptor blockade, by activating ETB receptors, increases vascular permeability and induces exaggerated fluid retention. J Pharmacol Exp Ther 361:322–333. 2017;

Article information Continued

Fig. 1.

Reclassification of six randomized clinical trials of clazosentan. *Among participants who were originally allocated in the clazosentan arm, those who received nimodipine concomitantly were not included in this post-hoc analysis. Participants who received the approved dose of 10 mg/h in Korean and Japan were included in this post-hoc analysis. In the nimodipine subgroup, participants were included if they received nimodipine 24 hours before the initiation of study treatment and up to 24 hours after its discontinuation. Participants who received nimodipine as a rescue medication were excluded from the nimodipine subgroup. RCT : randomized controlled trial.

Fig. 2.

Propensity score distributions. Propensity score density histogram of each subgroup used to analyze (A) moderate to severe angiographic vasospasm within 14 days after aSAH and (B) vasospasm-related morbidity and all-cause mortality within 6 weeks after aSAH. aSAH : aneurysmal subarachnoid hemorrhage.

Fig. 3.

Propensity score-matched analysis for angiographic vasospasm and vasospasm-related morbidity and all-cause mortality. Event rate (%) and relative risk reductions (95% CIs) are shown for (A) moderate to severe angiographic vasospasm within 14 days after aSAH and (B) vasospasm-related morbidity and all-cause mortality within 6 weeks after aSAH. The numbers on each bar represent the event rates (%) for each subgroup. Only statistically significant p-values based on Fisher’s exact test between two groups are displayed : otherwise, not significant. CI : confidence interval, aSAH : aneurysmal subarachnoid hemorrhage.

Table 1.

Summary of analyzed studies

AC-054-201 (CONSCIOUS-1) AC-054-202 AC-054-301 (CONSCIOUS-2) AC-054-302 (CONSCIOUS-3) AC-054-305 AC-054-306
Reference Macdonald et al. [22] (2008) Fujimura et al. [11] (2017) Macdonald et al. [19] (2011) Macdonald et al. [20] (2012) Endo et al. [10] (2022) Endo et al. [10] (2022)
Study region USA, Canada, Europe Korea & Japan USA, Canada, Europe, Asia USA, Canada, Europe, Asia, South America Japan Japan
Study period 2005–2006 2009–2010 2007–2010 2009–2011 2016–2020 2017–2020
Arms and numbers Placebo : 96 Placebo : 59 Placebo : 383 Placebo : 189 Placebo : 111 Placebo : 111
CLAZO 1 mg/h : 107 CLAZO 5 mg/h : 61 CLAZO 5 mg/h : 764 CLAZO 5 mg/h : 194 CLAZO 10 mg/h : 109 CLAZO 10 mg/h : 109
CLAZO 5 mg/h : 110 CLAZO 10 mg/h : 59 CLAZO 15 mg/h : 188
CLAZO 15 mg/h : 96
Nimodipine use (%) 86.0 19.0* 80.0 95.0 Not allowed* Not allowed*
Eligibility
 Age (years) 18–70 20–75 18–75 18–75 20–75 20–75
 Treatment procedure Clipping or coiling Clipping Clipping Coiling Coiling Clipping
 Fisher grade Not mentioned Fisher grade 3 Not mentioned Not mentioned Fisher grade 3 Fisher grade 3
 WFNS grade Grades I–IV, grade V patients who improved to ≤grade IV Grades I–IV Grades I–IV Grades I–IV Grades I–IV Grades I–IV
Clot size at baseline
 Diffuse thick 290 (71.4%) 166 (92.7%) 574 (50.1%) 345 (60.5%) 105 (47.7%) 118 (53.6%)
 Diffuse thin 103 (25.4%) 11 (6.1%) 149 (13.0%) 46 (8.1%) 54 (24.5%) 43 (19.5%)
 Local thick 10 (2.5%) 2 (1.1%) 395 (34.5%) 168 (29.5%) 17 (7.7%) 22 (10.0%)
 Local thin 3 (0.7%) 0 (0.0%) 25 (2.2%) 9 (1.6%) 31 (14.1%) 30 (13.6%)
No clot 1 (0.1%) 12 (5.5%) 7 (3.2%)
Endpoints & results
 Angiographic vasospasm Placebo : 66% Placebo : 80.0% - - Placebo : 49.5% Placebo : 55.0%
CLAZO 1 mg/h : 43% CLAZO 5 mg/h : 38.5% CLAZO 10 mg/h : 28.4% CLAZO 10 mg/h : 24.8%
CLAZO 5 mg/h : 39%
CLAZO 15 mg/h : 23% CLAZO 10 mg/h : 35.3%
 Vasospasm-related morbidity and all-cause mortality§ Placebo : 39% Placebo : 47.3% Placebo : 25% Placebo : 27% Placebo : 28.8% Placebo : 39.6%
CLAZO 1 mg/h : 37% CLAZO 5 mg/h : 28.8% CLAZO 5 mg/h : 21% CLAZO 5 mg/h : 24% CLAZO 10 mg/h : 13.6% CLAZO 10 mg/h : 16.2%
CLAZO 5 mg/h : 28% CLAZO 10 mg/h : 14.3% CLAZO 15 mg/h : 15%
CLAZO 15 mg/h : 29%
*

Nimodipine was allowed only for subjects enrolled from Korea in Study AC-504-202 as nimodipine is not approved in Japan.

Diffuse = long axis ≥20 mm; localized = long axis <20 mm; thick = short axis ≥4 mm; thin = 1 mm≤ short axis <4 mm; no clot = short axis <1 mm.

Determined by Image Review Committees (IRCs).

§

Centrally reviewed by expert committees; in Studies 305 and 306, cases of rescue therapy initiation were not included in vasospasmrelated morbidity events.

p-value <0.05 vs. placebo, Fisher’s exact test.

CONSCIOUS : clazosentan to overcome neurological ischemia and infarct occurring after subarachnoid hemorrhage, CLAZO : clazosentan, WFNS : World Federation of Neurosurgical Societies

Table 2.

Within-study comparison and subgroup pooled results of vasospasm and vasospasm-related morbidity and mortality

Clazosentan 10 mg/h Placebo Nimodipine Placebo
Analysis for angiographic vasospasm
 Study
  201 53/83 (63.86) 11/13 (84.62)
  202 21/49 (42.86) 35/45 (77.78) 13/14 (92.86) 35/45 (77.78)
  301
  302
  305 31/109 (28.44) 55/111 (49.55)
  306 27/109 (24.77) 61/111 (54.95)
 Total 79/267 (29.59) 151/267 (56.55) 66/97 (68.04) 46/58 (79.31)
 RRR (95% CI) 0.48 (0.35 to 0.58) 0.14 (-0.04 to 0.29)
Analysis for vasospasm-related morbidity and all-cause mortality
 Study
  201 35/83 (42.17) 5/13 (38.46)
  202 10/49 (20.41) 20/45 (44.44) 7/14 (50.00) 20/45 (44.44)
  301 80/327 (24.46) 17/56 (30.36)
  302 48/179 (26.82) 2/10 (20.00)
  305 23/109 (21.10) 34/111 (30.63)
  306 22/109 (20.18) 50/111 (45.05)
 Total 55/267 (20.60) 104/267 (38.95) 170/603 (28.19) 44/124 (35.48)
 RRR (95% CI) 0.47 (0.30 to 0.60) 0.21 (-0.04 to 0.39)

Values are presented as event/number of participants (%) unless otherwise indicated. RRR : relative risk reduction, CI : confidence interval

Table 3.

Baseline characteristics of participants in the propensity score-matched analysis

Analysis for angiographic vasospasm, Studies 201, 202, 305, and 306
Analysis for vasospasm-related morbidity and all-cause mortality, Studies 201, 202, 301, 302, 305, and 306
Clazosentan 10 mg/h Nimodipine Placebo Clazosentan 10 mg/h Nimodipine Placebo
Number 89 89 89 231 231 231
Sex
 Male 27 (30.3) 31 (34.8) 31 (34.8) 75 (32.5) 56 (24.2) 75 (32.5)
 Female 62 (69.7) 58 (65.2) 58 (65.2) 156 (67.5) 175 (75.8) 156 (67.5)
Age (years) 57.2±11.1 52.5±10.1 58.1±11.3 56.3±11.8 55.3±10.3 56.8±11.0
GCS score 13.1±2.6 13.3±2.7 13.7±2.1 13.2±2.4 13.2±2.4 13.3±2.2
Treatment procedure
 Coiling 38 (42.7) 44 (49.4) 40 (44.9) 91 (39.4) 89 (38.5) 87 (37.7)
 Clipping 51 (57.3) 45 (50.6) 49 (55.1) 140 (60.6) 142 (61.5) 144 (62.3)
WFNS grade
 Grade I 37 (41.6) 46 (51.7) 44 (49.4) 87 (37.7) 89 (38.5) 87 (37.7)
 Grade II 29 (32.6) 19 (21.3) 31 (34.8) 81 (35.1) 84 (36.4) 83 (35.9)
 Grade III 0 (0.0) 0 (0.0) 0 (0.0) 12 (5.2) 9 (3.9) 13 (5.6)
 Grade IV 23 (25.8) 24 (27.0) 14 (15.7) 51 (22.1) 49 (21.2) 48 (20.8)
Clot size
 Diffuse thick 53 (59.6) 70 (78.7) 52 (58.4) 143 (61.9) 151 (65.4) 146 (63.2)
 Other 36 (40.4) 19 (21.3) 37 (41.6) 88 (38.1) 80 (34.6) 85 (36.8)
Aneurysm location
 Anterior 83 (93.3) 68 (76.4) 79 (88.8) 215 (93.1) 178 (77.1) 215 (93.1)
 Posterior 6 (6.7) 16 (18.0) 7 (7.9) 15 (6.5) 51 (22.1) 13 (5.6)

Values are presented as mean±standard deviation or number (proportion). GCS : Glasgow coma scale, WFNS : World Federation of Neurosurgical Societies

Table 4.

Safety overview

Clazosentan all doses (n=479) Nimodipine (n=606) Placebo (n=347)
≥1 TEAE 446 (93.1) 553 (91.3) 326 (93.9)
 Treatment-related TEAE 137 (28.6) 126 (20.8) 54 (15.6)
Serious TEAE 114 (23.8) 210 (34.7) 68 (19.6)
 Treatment-related Serious TEAE 16 (3.3) 13 (2.1) 4 (1.2)
TEAE leading to treatment discontinuation 51 (10.6) 35 (5.8) 33 (9.5)
Main causes of death (up to 12 weeks post-aSAH) 24 (5.0) 28 (4.6) 13 (3.7)
 Cerebral infarction 7 (1.5) 16 (2.6) 4 (1.2)
 Cerebral ischaemia 5 (1.0) 1 (0.2) 0 (0.0)
 Cerebral vasoconstriction 4 (0.8) 13 (2.1) 7 (2.0)
 Brain oedema 4 (0.8) 4 (0.7) 1 (0.3)
 Acute kidney injury 1 (0.2) 2 (0.3) 0 (0.0)
 Subarachnoid haemorrhage 1 (0.2) 1 (0.2) 2 (0.6)
 Cerebral haemorrhage 1 (0.2) 1 (0.2) 0 (0.0)
 Multiple organ dysfunction syndrome 1 (0.2) 1 (0.2) 0 (0.0)
 Ruptured cerebral aneurysm 1 (0.2) 1 (0.2) 0 (0.0)
 Respiratory failure 1 (0.2) 0 (0.0) 1 (0.3)
 Blood pressure decreased 1 (0.2) 0 (0.0) 0 (0.0)
 Circulatory collapse 1 (0.2) 0 (0.0) 0 (0.0)
 Diabetes insipidus 1 (0.2) 0 (0.0) 0 (0.0)
 Haemorrhagic cerebral infarction 1 (0.2) 0 (0.0) 0 (0.0)
 Metabolic acidosis 1 (0.2) 0 (0.0) 0 (0.0)
 Sudden cardiac death 1 (0.2) 0 (0.0) 0 (0.0)
 Brain death 0 (0.0) 3 (0.5) 0 (0.0)
 Areflexia 0 (0.0) 1 (0.2) 0 (0.0)
 Brain herniation 0 (0.0) 1 (0.2) 0 (0.0)
 Carotid artery occlusion 0 (0.0) 1 (0.2) 0 (0.0)
 Cerebral haematoma 0 (0.0) 1 (0.2) 0 (0.0)
 Haemodynamic instability 0 (0.0) 1 (0.2) 0 (0.0)
 Hypotension 0 (0.0) 1 (0.2) 0 (0.0)
 Ileus paralytic 0 (0.0) 1 (0.2) 0 (0.0)
 Intraventricular haemorrhage 0 (0.0) 1 (0.2) 0 (0.0)
 Labile blood pressure 0 (0.0) 1 (0.2) 0 (0.0)
 Pneumonia aspiration 0 (0.0) 1 (0.2) 0 (0.0)
 Rhabdomyolysis 0 (0.0) 1 (0.2) 0 (0.0)
 Sepsis 0 (0.0) 1 (0.2) 0 (0.0)
 Acute respiratory failure 0 (0.0) 0 (0.0) 1 (0.3)
 Cardiac failure 0 (0.0) 0 (0.0) 1 (0.3)
 Pulmonary oedema 0 (0.0) 0 (0.0) 1 (0.3)

Values are presented as number (%). TEAEs were defined as adverse events from start of study treatment up to 24 hours after treatment discontinuation. TEAE : treatment-emergent adverse event, aSAH : aneurysmal subarachnoid hemorrhage

Table 5.

TEAEs reported by ≥3% of clazosentan-treated patients

Preferred term Clazosentan all doses (n=479) Nimodipine (n=606) Placebo (n=347)
Number of subjects with at least one TEAE 446 (93.1) 553 (91.3) 326 (93.9)
 Constipation 103 (21.5) 112 (18.5) 97 (28.0)
 Anaemia 96 (20.0) 66 (10.9) 40 (11.5)
 Pyrexia 91 (19.0) 132 (21.8) 75 (21.6)
 Hypokalaemia 88 (18.4) 82 (13.5) 47 (13.5)
 Cerebral vasoconstriction 80 (16.7) 189 (31.2) 127 (36.6)
 Hyponatraemia 78 (16.3) 114 (18.8) 59 (17.0)
 Delayed ischaemic neurological deficit 61 (12.7) 4 (0.7) 75 (21.6)
 Pleural effusion 61 (12.7) 26 (4.3) 12 (3.5)
 Headache 58 (12.1) 44 (7.3) 60 (17.3)
 Cerebral infarction 54 (11.3) 64 (10.6) 43 (12.4)
 Hypoalbuminaemia 49 (10.2) 14 (2.3) 33 (9.5)
 Brain oedema 42 (8.8) 45 (7.4) 15 (4.3)
 Pulmonary oedema 39 (8.1) 27 (4.5) 15 (4.3)
 Vomiting 37 (7.7) 30 (5.0) 22 (6.3)
 Pneumonia 36 (7.5) 65 (10.7) 13 (3.7)
 Cerebral ischaemia 29 (6.1) 14 (2.3) 12 (3.5)
 Hypotension 28 (5.8) 26 (4.3) 5 (1.4)
 Nausea 26 (5.4) 40 (6.6) 21 (6.1)
 Hepatic function abnormal 23 (4.8) 0 (0.0) 15 (4.3)
 Insomnia 22 (4.6) 11 (1.8) 22 (6.3)
 Gamma-glutamyltransferase increased 21 (4.4) 25 (4.1) 10 (2.9)
 Alanine aminotransferase increased 19 (4.0) 20 (3.3) 12 (3.5)
 Aspartate aminotransferase increased 18 (3.8) 13 (2.1) 8 (2.3)
 Urinary tract infection 17 (3.5) 110 (18.2) 15 (4.3)
 C-reactive protein increased 16 (3.3) 22 (3.6) 11 (3.2)
 Hepatic enzyme increased 16 (3.3) 43 (7.1) 16 (4.6)
 Hydrocephalus 15 (3.1) 46 (7.6) 18 (5.2)
 Hypertension 15 (3.1) 40 (6.6) 20 (5.8)
 Meningitis 15 (3.1) 16 (2.6) 18 (5.2)

Values are presented as number (%). TEAE : treatment-emergent adverse event