Skip to main content
Lippincott Open Access logoLink to Lippincott Open Access
. 2024 Sep 25;38(1):10–17. doi: 10.1097/WCO.0000000000001325

New targets in spontaneous intracerebral hemorrhage

Pu-Tien Chiang a,b, Li-Kai Tsai a, Hsin-Hsi Tsai a
PMCID: PMC11706352  PMID: 39325041

Abstract

Purpose of review

Intracerebral hemorrhage (ICH) is a devastating stroke with limited medical treatments; thus, timely exploration of emerging therapeutic targets is essential. This review focuses on the latest strategies to mitigate secondary brain injury post-ICH other than targeting surgery or hemostasis, addressing a significant gap in clinical practice and highlighting potential improvements in patient outcomes.

Recent findings

Promising therapeutic targets to reduce secondary brain injury following ICH have recently been identified, including attenuation of iron toxicity and inhibition of ferroptosis, enhancement of endogenous resorption of hematoma, and modulation of perihematomal inflammatory responses and edema. Additionally, novel insights suggest the lymphatic system of the brain may potentially play a role in hematoma clearance and edema management. Various experimental and early-phase clinical trials have demonstrated these approaches may potentially offer clinical benefits, though most research remains in the preliminary stages.

Summary

Continued research is essential to identify multifaceted treatment strategies for ICH. Clinical translation of these emerging targets could significantly enhance the efficacy of therapeutic interventions and potentially reduce secondary brain damage and improve neurological recovery. Future efforts should focus on large-scale clinical trials to validate these approaches, to pave the way for more effective treatment protocols for spontaneous ICH.

Keywords: ferroptosis, glymphatics, intracerebral hemorrhage, neuroinflammation, perihematomal edema

INTRODUCTION

Intracerebral hemorrhage (ICH) is a devastating form of stroke with a poor prognosis, and despite ongoing research, effective treatments remain limited. Stroke unit care benefits all ICH patients in general, though the individual effect is probably small. Hematoma volume and growth are key determinants of ICH outcomes, as early brain damage is primarily caused by the mass effect of the hematoma. Aggressive blood pressure control and hemostatic agents are currently considered the main treatment approaches for acute ICH, with modest effects on reducing mortality and dependency [1▪▪,2]. Minimally invasive surgery may be beneficial for a highly selected group of patients, particularly those with lobar ICH [3▪▪], but conventional surgery has not consistently shown benefit, underscoring the need for new treatment strategies.

After an ICH occurs, the blood products from the hematoma gradually lead to secondary brain injuries due to increased oxidative stress and related inflammatory responses [4]. Management of the consequences of secondary brain injuries has not been extensively investigated. Most of the related targets have only been investigated at the preclinical stage in experimental models of ICH. Some promising targets have been successfully translated to clinical trials but have only reached early-phase studies (Table 1); therefore, more research is needed to identify targets that may help to reduce secondary brain damage after an ICH occurs. 

Table 1.

Therapeutic targets in intracerebral hemorrhage that enter phase I/II trials

Drug n Study design Status ClinicalTrials. gov number Outcomes in completed trials
Deferoxamine (i-DEF) [28,35] 291 Phase II; randomized, double-blind, placebo-controlled Completed NCT02175225 No improvement of mRS score at day 90. Post hoc analysis suggested that drug accelerated and altered the trajectory of recovery up to 6 months
Pioglitazone (SHRINC) [12,13] 84 Phase II; randomized, double-blind, placebo-controlled Completed NCT00827892 No difference in mortality, cerebral edema, and functional outcomes
Remote ischemic conditioning (RICH) [15] 40 Phase I; randomized, open-label, evaluator-blind Completed NCT03930940 No difference in hematoma size and absolute PHE at day 7, but higher hematoma resolution rate and lower relative PHE in RIC group were observed
Minocycline (MACH) [22] 16 Phase I/II; randomized, open-label, evaluator-blind Completed NCT01805895 No difference in mRS, inflammatory biomarkers, hematoma volume, or PHE at 90 days
Minocycline (MITCH) [23] 20 Phase I/II; randomized, double-blind, placebo-controlled Completed NCT03040128 No difference in clinical and radiological outcomes. Drug was associated with a decrease in MMP-9 level
Minocycline (BATMAN) [27] Phase I/II; randomized, double-blind, placebo-controlled Recruiting NCT05680389
Anakinra (BLOC-ICH) [29] 25 Phase II; randomized, double-blind, placebo-controlled Completed NCT03737344 No difference in oedema extension distance and mRS at 3 months. Post hoc analysis indicated reduced IL-6 levels with anakinra
Anakinra (ACTION) [30] 75 Phase II; randomized, open-label, evaluator-blind, three-armed Recruiting NCT04834388
Fingolimod (FITCH) 28 Phase I; randomized, double-blind, placebo-controlled Completed NCT04088630 Pending
Siponimod [42] 32 Phase II; randomized, double-blind, placebo-controlled Completed NCT03338998 No difference in absolute PHE volume
Glibenclamide (GATE-ICH) [66] 200 Phase II; randomized, open-label, evaluator-blind Completed NCT03741530 Drug did not improve functional outcome at day 90
Celecoxib 60 Phase II; randomized, open-label Recruiting NCT05434065
Statin discontinuation vs. continuation (SATURN) [77] 1456 Phase III; randomized, open-label, evaluator-blind Recruiting NCT03936361
Atorvastatin (STATIC) [78] 98 Phase II/III; randomized, open-label, evaluator-blind Recruiting NCT04857632
Conivaptan [83] 7 Phase I; open-label Completed NCT03000283 Drug was well tolerated, with no change in cardiac or renal function

IL, interleukin; MMP, matrix metalloproteinase; mRS, modified Rankin Scale; PHE, perihematomal edema; RIC, remote ischemic conditioning.

Box 1.

Box 1

no caption available

This review summaries the recent advances for various therapeutic targets that may reduce secondary brain injury after ICH, including reduction of toxicity from blood components, promotion of endogenous hematoma resorption, modulation of perihematomal inflammatory responses, and alleviation of perihematomal edema (PHE). We also discuss the emerging knowledge on the role of the lymphatic system, which may participate in hematoma clearance and contribute to the formation of PHE after ICH. Important trials of these novel therapeutic targets are summarized in Table 1.

IRON TOXICITY AND FERROPTOSIS

After an ICH occurs, neurotoxins from the hematoma, such as hemoglobin and heme, are released into the brain parenchyma and metabolized into ferrous (Fe2+) or ferric ion (Fe3+) [5]. This process induces iron toxicity that involves the overproduction of reactive oxygen species (ROS) and iron-dependent nonapoptotic cell death, also called ferroptosis [6,7]. Ferroposis is a form of programmed cell death that is characterized by morphological changes in mitochondria and iron-dependent lipid peroxidation [7,8].

Attenuation of iron toxicity or inhibition of ferroptosis has been considered as a potential therapeutic target to reduce secondary brain injury following ICH. Infusion of the iron chelator agent deferoxamine mesylate within 24 h of the onset of ICH was investigated in a phase II clinical trial (i.e. deferoxamine mesylate in patients with ICH, iDEF trial), which confirmed this approach was well tolerated but potentially had low efficacy in terms of 90-day functional outcomes [9]. However, a more recently published post hoc analysis of the iDEF trial for long-term recovery suggested that deferoxamine accelerated recovery and altered the trajectory of functional outcomes in the 6 months after ICH [10].

Targets that inhibit heme-induced ferroptosis have been found to have beneficial effects after ICH; however, most of these findings were based on preclinical studies [11]. Increased expression of heme oxygenase-1 (HO-1), which catalyzes heme to release Fe2+, is observed after ICH, especially in the peri-hematoma area [12,13]. Recent advances suggest that targeting pathways that alter HO-1-related signaling could effectively attenuate ferroptosis-induced brain injury [1416]. However, further research is required to determine how ferroptosis interacts with other traditional cell death pathways and elucidate its contribution to brain injury [17].

ENDOGENOUS HEMATOMA RESORPTION

Currently, minimally invasive removal of the hematoma is the primary treatment approach for rapid hematoma clearance; however, this treatment is restricted to patients with hematoma located in the lobar regions and is only effective within the early time window [3▪▪,18]. In addition, surgical approaches are more frequently employed for patients with larger hematoma, and therefore the role of surgery in minor ICH remains uncertain. Acceleration of endogenous absorption of hematoma has been suggested as a potential strategy to reduce secondary brain injury due to hemolytic blood products, especially for patients who are ineligible for surgical hematoma removal.

Erythrophagocytosis by microglia/macrophages is an important endogenous mechanism that removes red blood cells, detoxifies hemolytic products, and facilitates neurological recovery [19]. Enhancement of peroxisome proliferator-activated receptor (PPAR)-γ or NRF2-related signaling though controlling the expression of scavenger receptor genes, which promotes erythrophagocytosis by microglia/macrophages, has been found to increase hematoma resorption [2022]. Bexarotene, a retinoid X receptor agonist that may promote activation of PPAR-γ, and vitamin D have been found to potentially enhance hematoma clearance in experimental ICH models [23,24]. Drugs targeting PPAR-γ or related pathways may potentially improve patient outcomes; however, a phase II clinical trial of pioglitazone, a potent PPAR-γ agonist, demonstrated no difference in mortality, cerebral edema, or functional outcomes (SHRINC, n = 84) [25,26].

Remote ischemic conditioning (RIC), a maneuver that involves repetitive inflation–deflation of a blood pressure cuff on a limb, has been shown to promote clot resolution and attenuate cerebral edema in an animal model via AMPK-dependent immune regulation [27]. Repeated daily RIC for 7 days was evaluated in a phase I trial (RICH, n = 40) and was found to be well tolerated in patients with ICH. It may potentially improve the rate of hematoma resolution [28].

INFLAMMATORY RESPONSES

Following an ICH, various inflammatory pathways are activated around the hematoma. These processes involve activation of microglia and astrocytes, as well as leukocyte infiltration. The resulting neuroinflammation, which begins within hours and persists for weeks, provides a window for therapeutic intervention [29,30,31].

Microglia and macrophages exist in either a proinflammatory (M1) state, producing cytokines like TNF-α and IL-1β, or a regulatory (M2) state, releasing anti-inflammatory cytokines such as TGF-β and IL-10 [29,30]. Minocycline, a second-generation tetracycline, has been reported to exert several nonantimicrobial properties that may be beneficial for ICH treatment, including inhibition of pro-inflammatory microglia/macrophages and matrix metalloproteases (MMPs), antioxidant effects, and reduction of neuronal apoptotic signaling [3234]. Small phase I/II trials (MACH [35], n = 16; MITCH [36], n = 20) reported that minocycline reduced serum MMP-9 levels in patients with ICH, although no significant differences in clinical outcomes were observed. A retrospective cohort study of patients with clinically aggressive cerebral amyloid angiopathy (CAA) demonstrated that minocycline was associated with a reduced risk of ICH recurrence [37]. A phase I/II study of minocycline for CAA is currently underway (BATMAN, NCT05680389) [38].

Cytokines play pivotal roles in the polarization and functional regulation of microglia. Anakinra, a recombinant human IL-1 receptor antagonist, was evaluated in a phase II study (BLOC-ICH [39▪▪], n = 25); although no significant improvements in radiological or clinical outcomes were observed, a post hoc analysis indicated reduced IL-6 levels with anakinra at day 2 after ICH. Another phase II trial of Anakinra (ACTION [40], NCT04834388) is currently ongoing. TGF-β1 modulates microglia-mediated neuroinflammation and supports functional recovery, as demonstrated in animal studies, and therefore may also have therapeutic potential for ICH [41,42].

Sphingosine-1-phosphate (S1P) is a bioactive lipid mediator that acts through the S1P receptors (S1PR1–S1PR5) [30,43]. Activation of the S1P receptors inhibits the egress of lymphocytes from lymph nodes and their recirculation [44], and also appears to reduce pro-inflammatory responses and enhance regulatory functions in microglia [45]. Fingolimod, an S1PR agonist, was demonstrated to reduce PHE and neurological deficits in a small proof-of-concept clinical study (n = 23) [46]. A phase I study of fingolimod (FITCH, NCT04088630) in 28 patients with ICH has been completed; however, the results are pending. Siponimod, another S1PR modulator, was tested in a phase II trial (NCT03338998) that was halted because of the COVID-19 pandemic, with no significant differences in edema volume observed in the final 29 patients [47].

Moreover, ICH activates the Nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, which amplifies inflammation by promoting the release of IL-1β and neutrophil infiltration, which in turn worsen brain edema [48]. In animal models, inhibition of NLRP3 reduced cerebral edema and neurological deficits [49,50,51]. The 18 kDa translocator protein (TSPO), which is upregulated in peri-hematomal regions post-ICH, is primarily observed in microglia/macrophages and regulates the release of pro-inflammatory cytokines [52]. TSPO ligands such as etifoxine have been shown to reduce neurodeficits and brain edema in animal models [53]. In addition, the complement system is activated at an early stage in ICH, and triggers microglial activation and subsequent neuroinflammation [54]. C3a and C5a receptor antagonists have been found to improve brain edema and neurological function [54], and the complement inhibitor N-acetyl heparin (NAH) reduced brain injury and iron accumulation postintraventricular hemorrhage in animal models [55].

Reactive astrocytes, which are classified as the proinflammatory A1 and reparative A2 subtypes, also play crucial roles in neuroinflammation [31]. The aquaporin-4 (AQP4) water channel protein is located in astrocyte foot processes and is essential for maintenance of blood–brain barrier integrity. Enhancing the expression of AQP4 reduced PHE [56]. Moreover, edaravone, which maintains the polarity of AQP4, alleviated brain edema and injury [57]. IL-15, which colocalizes with astrocytes, exacerbated neurological deficits and cerebral edema and modulated microglia towards a pro-inflammatory phenotype in GFAP-IL-15tg mice [58]. Conversely, IL-33, which is expressed in astrocytes, suppressed pro-inflammatory cytokines, reduced cerebral water content, and improved neurological function post-ICH in mice ICH model [59].

Neutrophil-driven inflammation and cytotoxicity are generally believed to worsen ICH injury. IL-27, which is produced by microglia/macrophages, increased production of lactoferrin by neutrophils and reduced brain edema, neuronal death, and functional deficits [60]. Lastly, activation of the PD-1/PD-L1 pathway, which inhibits T-cell activity, reduced neurologic deficits, cerebral edema, and the hemorrhage volume in ICH [61], and also potentially shifted microglia to an anti-inflammatory phenotype [62].

PERIHEMATOMAL EDEMA

PHE is a significant secondary injury after ICH that contributes to the mass effect and increased intracranial pressure [63]. The standard management of cerebral edema includes osmolar therapies such as mannitol or hypertonic saline [63]. Thrombin, which breaks down the blood–brain barrier and promotes inflammation, is a key factor in brain edema [64]. In a rat ICH model, inhibition of thrombin using argatroban reduced secondary brain damage, including edema and inflammation [65]. However, thrombin is a crucial component of the coagulation cascade, and thus is a challenging therapeutic target for ICH.

Glibenclamide (glyburide), a sulfonylurea, inhibits Sur1-TRMP4 channels and has been shown to reduce PHE in animal models [66]. For ischemic stroke, a phase II trial (GAMES-RP [67], n = 77) of patients with large hemispheric infarcts showed glyburide reduced cerebral edema and mortality due to edema compared with the placebo, despite both groups achieving similar functional outcomes. The terminated phase III CHARM trial (NCT02864953, n = 585) of glibenclamide reported no overall functional improvement at 3 months [68], though patients with stroke volumes 120 ml or less had favorable outcomes in subgroup analysis [69]. For hemorrhagic stroke, the phase II GATE-ICH trial (n = 200) of patients with ICH found glibenclamide had no functional benefit at day 90 and was associated with a higher incidence of hypoglycemia [70], whereas the phase II ASTRAL trial (NCT03954041, n = 92) in patients with brain contusion showed no significant difference in contusion volume or functional outcomes [71].

Cyclo-oxygenase 2 (COX2) expression increases following ICH [72]. Celecoxib, a selective COX2 inhibitor, was found to reduce ICH-induced edema in animal models and in a case–control study [73,74]. The ACE-ICH pilot study (n = 44) indicated that treatment with celecoxib within 24 h was associated with reduced PHE expansion within the first 7 days post-ICH [75]. A phase II clinical trial (NCT05434065) is currently investigating the effects of administering celecoxib within 6 h of the onset of ICH.

Statins inhibit 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase and exert anti-inflammatory properties. In rodent ICH models, statins reduced PHE, decreased blood–brain barrier permeability, and improved functional outcomes and mortality [76]. Retrospective studies in humans have reported mixed results regarding prior statin use and PHE [77,78]. Analysis of a single-center prospective registry that included 1275 patients found initiation of statins post-ICH was linked to larger peak cerebral edema, while the continuation of prior statin use was not associated with a significant difference in peak edema compared with discontinuation [79]. Current trials investigating the role of statins in ICH include the phase III SATURN (NCT03936361) and phase II/III STATIC (NCT04857632) trials [80,81].

Vasopressin receptor antagonists, such as conivaptan and tolvaptan, increase plasma osmolarity and sodium levels. In experimental models of ICH, tolvaptan reduced brain edema and improved functional outcomes [82,83]. An open-label phase I trial of seven patients with ICH found conivaptan was well tolerated and did not lead to adverse cardiac or renal effects [84]. However, sodium overcorrection, hypotension, and hypokalemia were observed in another cohort study of patients treated with conivaptan or tolvaptan in neurocritical care [85].

EMERGING TARGET: THE BRAIN LYMPHATIC SYSTEM

The lymphatic system of the brain, including the glymphatics and meningeal lymphatics, plays a pivotal role in brain clearance of solutes, interstitial fluid, and metabolic waste [86]. As in other central nervous system (CNS) disorders, the lymphatic system has emerged as new target to alleviate secondary brain injury and to improve neurological recovery after ICH.

Perivascular spaces, which are surrounded by astrocytic endfeet, are key structures involved in cerebrospinal fluid (CSF) and interstitial fluid exchange and therefore regulate the function of the glymphatics [87]. Previous studies have suggested an association between enlarged PVS and increased risk of ICH recurrence, which implies a pathogenic role for the glymphatics in hemorrhagic stroke [88,89]. Within the PVS, AQP4 water channels expressed in the astrocytic endfeet have been investigated as potential targets to modulate water movement and the function of the glymphatics in ICH. Upregulation of AQP4 may protect the blood–brain barrier and help to resolve PHE (as mentioned above), whereas downregulation of AQP4 was associated with glymphatic dysfunction and had detrimental effects on neurological deficits following ICH in more recent studies [60,61,90].

The discovery of the meningeal lymphatic vessels (mLVs) generated broad research interest and has prompted investigation of their roles in various CNS diseases, including ICH. The mLVs are embedded within the dura mater and the vessel branches extend into the subarachnoid space, where they make direct contact with the CSF, and carry macromolecules away from the brain parenchyma and transport CSF to the cervical lymph nodes [91]. One study demonstrated the meningeal lymphatic system was involved in hematoma resolution, red blood cell drainage, and neurological recovery in an animal model of ICH [92]. Additionally, cilostazol, a drug that has been found to promote lymphatic flow, improved lymphatic function and hematoma clearance in an experimental model [92]. Therefore, targeting the lymphatic systems within the brain, including the glymphatics or meningeal lymphatics, may hold substantial therapeutic potential; however, more research is needed before these findings can be translated into clinical treatments.

CONCLUSION

The exploration of new therapeutic targets for spontaneous ICH is both timely and necessary, given the current lack of effective medical treatments. Recent advancements in our understanding of how to mitigate secondary brain injury post-ICH, such as reducing iron toxicity, enhancing endogenous resorption of hematoma, modulating inflammatory responses, and addressing PHE, offer promising avenues to improve patient outcomes. The lymphatic system of the brain also presents a novel area for therapeutic intervention. Despite the promising results obtained in experimental model and early-phase trials, further large-scale studies are essential to validate and translate these novel therapeutic targets into clinical practice. Continued research and innovation in this field are necessary to develop effective management protocols and ultimately improve the prognosis of patients with ICH.

Acknowledgements

None.

Financial support and sponsorship

This article is supported by funding from the National Science and Technology Council (Tsai HH, 113-2628-B-002-013-MY3 and 112-2923-B-002-001-MY3).

Conflicts of interest

There are no conflicts of interest.

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • ▪ of special interest

  • ▪▪ of outstanding interest

REFERENCES

  • 1▪▪.Ma L, Hu X, Song L, et al. INTERACT3 Investigators. The third Intensive Care Bundle with Blood Pressure Reduction in Acute Cerebral Haemorrhage Trial (INTERACT3): an international, stepped wedge cluster randomised controlled trial. Lancet 2023; 402:27–40. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates significant improvements in functional recovery and reduced adverse events in acute ICH patients through a care bundle protocol. The results support its integration into clinical practice, particularly in high-incidence settings.
  • 2.Greenberg SM, Ziai WC, Cordonnier C, et al. American Heart Association/American Stroke Association. 2022 Guideline for the management of patients with spontaneous intracerebral hemorrhage: a guideline from the American Heart Association/American Stroke Association. Stroke 2022; 53:e282–e361. [DOI] [PubMed] [Google Scholar]
  • 3▪▪.Pradilla G, Ratcliff JJ, Hall AJ, et al. ENRICH trial investigators, ENRICH Trial Investigators. Trial of early minimally invasive removal of intracerebral hemorrhage. New Engl J Med 2024; 390:1277–1289. [DOI] [PubMed] [Google Scholar]; This study demonstrates that early minimally invasive hematoma removal significantly improves functional outcomes at 180 days compared with medical management alone. It emphasizes the potential of minimally invasive surgery for improving patient recovery, especially in lobar ICH.
  • 4.Aronowski J, Zhao X. Molecular pathophysiology of cerebral hemorrhage. Stroke 2011; 42:1781–1786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wan J, Ren H, Wang J. Iron toxicity, lipid peroxidation and ferroptosis after intracerebral haemorrhage. Stroke Vasc Neurol 2019; 4:93–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Li Q, Han X, Lan X, et al. Inhibition of neuronal ferroptosis protects hemorrhagic brain. JCI Insight 2017; 2:e90777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bai Q, Liu J, Wang G. Ferroptosis, a regulated neuronal cell death type after intracerebral hemorrhage. Front Cell Neurosci 2020; 14:591874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lu C, Tan C, Ouyang H, et al. Ferroptosis in intracerebral hemorrhage: a panoramic perspective of the metabolism, mechanism and theranostics. Aging Dis 2022; 13:1348–1364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Selim M, Foster LD, Moy CS, et al. i-DEF Investigators. Deferoxamine mesylate in patients with intracerebral haemorrhage (i-DEF): a multicentre, randomised, placebo-controlled, double-blind phase 2 trial. Lancet Neurol 2019; 18:428–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Foster L, Robinson L, Yeatts SD, et al. i-DEF Investigators. Effect of deferoxamine on trajectory of recovery after intracerebral hemorrhage: a post hoc analysis of the i-DEF Trial. Stroke 2022; 53:2204–2210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Pan F, Xu W, Ding J, et al. Elucidating the progress and impact of ferroptosis in hemorrhagic stroke. Front Cell Neurosci 2023; 16:1067570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Puy L, Perbet R, Figeac M, et al. Brain peri-hematomal area, a strategic interface for blood clearance: a human neuropathological and transcriptomic study. Stroke 2022; 53:2026–2035. [DOI] [PubMed] [Google Scholar]
  • 13.Li QQ, Li LJ, Wang XY, et al. Research progress in understanding the relationship between heme oxygenase-1 and intracerebral hemorrhage. Front Neurol 2018; 9:682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14▪.Wang R, Liang Z, Xue X, et al. Microglial FoxO3a deficiency ameliorates ferroptosis-induced brain injury of intracerebral haemorrhage via regulating autophagy and heme oxygenase-1. J Cell Mol Med 2024; 28:e18007. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study highlights the potential of targeting FoxO3a to reduce neuroinflammation in ICH. It suggests that regulating autophagy and heme oxygenase-1 can mitigate ferroptosis-induced brain injury.
  • 15▪.Zhou ZX, Cui Q, Zhang YM, et al. Withaferin A inhibits ferroptosis and protects against intracerebral hemorrhage. Neural Regen Res 2023; 18:1308. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates Withaferin A's potential to reduce oxidative stress and ferroptosis in ICH. The findings highlight the role of the Nrf2/HO-1-signaling pathway in this neuroprotection.
  • 16▪.Jiang Z, Yang H, Ni W, et al. Attenuation of neuronal ferroptosis in intracerebral hemorrhage by inhibiting HDAC1/2: Microglial heterogenization via the Nrf2/HO1 pathway. CNS Neurosci Ther 2024; 30:e14646. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that inhibiting HDAC1/2 can reduce neuronal ferroptosis in ICH by modulating microglial heterogenization. It highlights the Nrf2/HO1 pathway as a crucial mechanism in this neuroprotection.
  • 17.Cao Y, Xiao W, Liu S, et al. Ferroptosis: Underlying mechanism and the crosstalk with other modes of neuronal death after intracerebral hemorrhage. Front Cell Neurosci 2023; 17:1080344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mendelow AD, Gregson BA, Rowan EN, et al. STICH II Investigators. Early surgery versus initial conservative treatment in patients with spontaneous supratentorial lobar intracerebral haematomas (STICH II): a randomised trial. Lancet 2013; 382:397–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Liu J, Zhu Z, Leung GKK. Erythrophagocytosis by microglia/macrophage in intracerebral hemorrhage: from mechanisms to translation. Front Cell Neurosci 2022; 16:818602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wang G, Li T, Duan SN, et al. PPAR-γ promotes hematoma clearance through haptoglobin-hemoglobin-CD163 in a rat model of intracerebral hemorrhage. Behav Neurol 2018; 2018:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zhao X, Sun G, Ting S, et al. Cleaning up after ICH: the role of Nrf2 in modulating microglia function and hematoma clearance. J Neurochem 2015; 133:144–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.You M, Long C, Wan Y, et al. Neuron derived fractalkine promotes microglia to absorb hematoma via CD163/HO-1 after intracerebral hemorrhage. Cell Mol Life Sci 2022; 79:224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chang CF, Massey J, Osherov A, et al. Bexarotene enhances macrophage erythrophagocytosis and hematoma clearance in experimental intracerebral hemorrhage. Stroke 2020; 51:612–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Liu J, Li N, Zhu Z, et al. Vitamin D enhances hematoma clearance and neurologic recovery in intracerebral hemorrhage. Stroke 2022; 53:2058–2068. [DOI] [PubMed] [Google Scholar]
  • 25.Gonzales NR, Shah J, Sangha N, et al. Design of a prospective, dose-escalation study evaluating the Safety of Pioglitazone for Hematoma Resolution in Intracerebral Hemorrhage (SHRINC). Int J Stroke 2013; 8:388–396. [DOI] [PubMed] [Google Scholar]
  • 26.Gonzales N, Sangha N, Cai C, et al. The Safety of Pioglitazone for Hematoma Resolution in IntraCerebral Hemorrhage (SHRINC): a randomised, blinded, phase 2, safety trial. SSRN Electronic J 2018; [Epub ahead of print]. [Google Scholar]
  • 27.Vaibhav K, Braun M, Khan MB, et al. Remote ischemic postconditioning promotes hematoma resolution via AMPK-dependent immune regulation. J Exp Med 2018; 215:2636–2654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Zhao W, Jiang F, Li S, et al. RICH-1 Investigators. Safety and efficacy of remote ischemic conditioning for the treatment of intracerebral hemorrhage: a proof-of-concept randomized controlled trial. Int J Stroke 2022; 17:425–433. [DOI] [PubMed] [Google Scholar]
  • 29.Tschoe C, Bushnell CD, Duncan PW, et al. Neuroinflammation after intracerebral hemorrhage and potential therapeutic targets. J Stroke 2020; 22:29–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bai Q, Xue M, Yong VW. Microglia and macrophage phenotypes in intracerebral haemorrhage injury: therapeutic opportunities. Brain 2020; 143:1297–1314. [DOI] [PubMed] [Google Scholar]
  • 31.Xue M, Yong VW. Neuroinflammation in intracerebral haemorrhage: immunotherapies with potential for translation. Lancet Neurol 2020; 19:1023–1032. [DOI] [PubMed] [Google Scholar]
  • 32.Wasserman JK, Zhu X, Schlichter LC. Evolution of the inflammatory response in the brain following intracerebral hemorrhage and effects of delayed minocycline treatment. Brain Res 2007; 1180:140–154. [DOI] [PubMed] [Google Scholar]
  • 33.Kobayashi K, Imagama S, Ohgomori T, et al. Minocycline selectively inhibits M1 polarization of microglia. Cell Death Dis 2013; 4:e525–e1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Naderi Y, Panahi Y, Barreto G, et al. Neuroprotective effects of minocycline on focal cerebral ischemia injury: a systematic review. Neural Regen Res 2020; 15:773–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Fouda AY, Newsome AS, Spellicy S, et al. Minocycline in acute cerebral hemorrhage an early phase randomized trial. Stroke 2017; 48:2885–2887. [DOI] [PubMed] [Google Scholar]
  • 36.Chang JJ, Kim-Tenser M, Emanuel BA, et al. Minocycline and matrix metalloproteinase inhibition in acute intracerebral hemorrhage: a pilot study. Eur J Neurol 2017; 24:1384–1391. [DOI] [PubMed] [Google Scholar]
  • 37▪.Bax F, Warren A, Fouks AA, et al. Minocycline in severe cerebral amyloid angiopathy: a single-center cohort study. J Am Heart Assoc 2024; 13:e033464. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study suggests minocycline may reduce the recurrence of intracerebral haemorrhage in severe cerebral amyloid angiopathy. It highlights minocycline's potential safety and tolerability in this patient population.
  • 38▪.Voigt S, Koemans EA, Rasing I, et al. Minocycline for sporadic and hereditary cerebral amyloid angiopathy (BATMAN): study protocol for a placebo-controlled randomized double-blind trial. Trials 2023; 24:378. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study aims to investigate minocycline's potential to reduce neuroinflammation in CAA. The trial assesses CSF biomarkers and MRI markers to evaluate treatment efficacy.
  • 39▪▪.Parry-Jones AR, Stocking K, MacLeod MJ, et al. Phase II randomised, placebo-controlled, clinical trial of interleukin-1 receptor antagonist in intracerebral haemorrhage: BLOcking the Cytokine IL-1 in ICH (BLOC-ICH). Eur Stroke J 2023; 8:819–827. [DOI] [PMC free article] [PubMed] [Google Scholar]; This phase II trial demonstrates the feasibility and preliminary safety of delivering anakinra in patients with acute ICH. Although no significant differences in perihematomal oedema or clinical outcomes were observed, a post hoc analysis indicated reduced IL-6 levels with anakinra.
  • 40▪.Cliteur MP, van der Kolk AG, Hannink G, et al. Anakinra in cerebral haemorrhage to target secondary injury resulting from neuroinflammation (ACTION): Study protocol of a phase II randomised clinical trial. Eur Stroke J 2024; 9:265–273. [DOI] [PMC free article] [PubMed] [Google Scholar]; This phase II trial aims to investigate the effect of anakinra on PHE in ICH patients, comparing high and low doses, and determine if anakinra reduces secondary brain injury and inflammation markers.
  • 41.Taylor RA, Chang CF, Goods BA, et al. TGF-β1 modulates microglial phenotype and promotes recovery after intracerebral hemorrhage. J Clin Invest 2017; 127:280–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42▪.Wen H, Tan J, Tian M, et al. TGF-β1 ameliorates BBB injury and improves long-term outcomes in mice after ICH. Biochem Biophys Res Commun 2023; 654:136–144. [DOI] [PubMed] [Google Scholar]; This study demonstrates the potential of TGF-β1 in reducing BBB injury in ICH mice. It also highlights its role in promoting long-term neurological recovery.
  • 43.O'Sullivan S, Dev KK. Sphingosine-1-phosphate receptor therapies: advances in clinical trials for CNS-related diseases. Neuropharmacology 2017; 113 (Pt b):597–607. [DOI] [PubMed] [Google Scholar]
  • 44.Rosen H, Stevens RC, Hanson M, et al. Sphingosine-1-phosphate and its receptors: structure, signaling, and influence. Annu Rev Biochem 2013; 82:637–662. [DOI] [PubMed] [Google Scholar]
  • 45.Qin C, Fan WH, Liu Q, et al. Fingolimod protects against ischemic white matter damage by modulating microglia toward M2 polarization via STAT3 pathway. Stroke 2017; 48:3336–3346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fu Y, Hao J, Zhang N, et al. Fingolimod for the treatment of intracerebral hemorrhage: a 2-arm proof-of-concept study. JAMA Neurol 2014; 71:1092–1101. [DOI] [PubMed] [Google Scholar]
  • 47.A phase II, patient- and investigator-blinded, randomized, placebo-controlled study to evaluate efficacy, safety and tolerability of BAF312 (siponimod) in patients with stroke due to intracerebral hemorrhage (ICH). Available at: https://www.novctrd.com/ctrdweb/trialresult/trialresults/pdf?trialResultId=17824. [Accessed 17 July 2024]. [Google Scholar]
  • 48.Ma Q, Chen S, Hu Q, et al. NLRP3 inflammasome contributes to inflammation after intracerebral hemorrhage. Ann Neurol 2014; 75:209–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49▪.Fang M, Xia F, Wang J, et al. The NLRP3 inhibitor, OLT1177 attenuates brain injury in experimental intracerebral hemorrhage. Int Immunopharmacol 2024; 131:111869. [DOI] [PubMed] [Google Scholar]; This study demonstrates that OLT1177 significantly reduces brain oedema and neurological deficits in ICH. It highlights the potential of NLRP3 inhibition in protecting against ICH-induced brain injury.
  • 50.Ren H, Kong Y, Liu Z, et al. Selective NLRP3 (pyrin domain–containing protein 3) inflammasome inhibitor reduces brain injury after intracerebral hemorrhage. Stroke 2018; 49:184–192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Yang X, Sun J, Kim TJ, et al. Pretreatment with low-dose fimasartan ameliorates NLRP3 inflammasome-mediated neuroinflammation and brain injury after intracerebral hemorrhage. Exp Neurol 2018; 310:22–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Bonsack F, Alleyne CH, Sukumari-Ramesh S. Augmented expression of TSPO after intracerebral hemorrhage: a role in inflammation? J Neuroinflammation 2016; 13:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Li M, Ren H, Sheth KN, et al. A TSPO ligand attenuates brain injury after intracerebral hemorrhage. FASEB J 2017; 31:3278–3287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Li Y, Tao C, An N, et al. Revisiting the role of the complement system in intracerebral hemorrhage and therapeutic prospects. Int Immunopharmacol 2023; 123:110744. [DOI] [PubMed] [Google Scholar]
  • 55▪.Zhang T, Xia F, Wan Y, et al. Complement inhibition reduces early erythrolysis, attenuates brain injury, hydrocephalus, and iron accumulation after intraventricular hemorrhage in aged rats. Transl Stroke Res 2024; [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]; This study shows that complement inhibition significantly reduces brain injury and hydrocephalus after IVH. The findings support its potential as a therapeutic target.
  • 56.Jeon H, Kim M, Park W, et al. Upregulation of AQP4 improves blood–brain barrier integrity and perihematomal edema following intracerebral hemorrhage. Neurotherapeutics 2021; 18:2692–2706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57▪.Wang Z, Li Y, Wang Z, et al. Edaravone maintains AQP4 polarity via OS/MMP9/β-DG pathway in an experimental intracerebral hemorrhage mouse model. Mol Neurobiol 2024; [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]; This study highlights edaravone's role in preserving AQP4 polarity and reducing brain edema in ICH. It underscores the importance of the OS/MMP9/β-DG pathway in neuroprotection.
  • 58.Shi SX, Li YJ, Shi K, et al. IL (Interleukin)-15 bridges astrocyte-microglia crosstalk and exacerbates brain injury following intracerebral hemorrhage. Stroke 2020; 51:967–974. [DOI] [PubMed] [Google Scholar]
  • 59.Gao Y, Ma L, Luo CL, et al. IL-33 exerts neuroprotective effect in mice intracerebral hemorrhage model through suppressing inflammation/apoptotic/autophagic pathway. Mol Neurobiol 2017; 54:3879–3892. [DOI] [PubMed] [Google Scholar]
  • 60.Zhao X, Ting SM, Liu CH, et al. Neutrophil polarization by IL-27 as a therapeutic target for intracerebral hemorrhage. Nat Commun 2017; 8:602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Han R, Luo J, Shi Y, et al. PD-L1 (programmed death ligand 1) protects against experimental intracerebral hemorrhage-induced brain injury. Stroke 2017; 48:2255–2262. [DOI] [PubMed] [Google Scholar]
  • 62.Wu J, Sun L, Li H, et al. Roles of programmed death protein 1/programmed death-ligand 1 in secondary brain injury after intracerebral hemorrhage in rats: selective modulation of microglia polarization to anti-inflammatory phenotype. J Neuroinflammation 2017; 14:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wan Y, Holste KG, Hua Y, et al. Brain edema formation and therapy after intracerebral hemorrhage. Neurobiol Dis 2023; 176:105948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lee KR, Kawai N, Kim S, et al. Mechanisms of edema formation after intracerebral hemorrhage: effects of thrombin on cerebral blood flow, blood-brain barrier permeability, and cell survival in a rat model. J Neurosurg 1997; 86:272–278. [DOI] [PubMed] [Google Scholar]
  • 65.Nagatsuna T, Nomura S, Suehiro E, et al. Systemic administration of argatroban reduces secondary brain damage in a rat model of intracerebral hemorrhage: histopathological assessment. Cerebrovasc Dis 2005; 19:192–200. [DOI] [PubMed] [Google Scholar]
  • 66.Jiang B, Li L, Chen Q, et al. Role of glibenclamide in brain injury after intracerebral hemorrhage. Transl Stroke Res 2017; 8:183–193. [DOI] [PubMed] [Google Scholar]
  • 67.Kimberly WT, Bevers MB, Von Kummer R, et al. Effect of IV glyburide on adjudicated edema endpoints in the GAMES-RP Trial. Neurology 2018; 91:e2163–e2169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.INTRAVENOUS GLIBENCLAMIDE FOR LARGE HEMISPHERIC INFARCTION: RESULTS FROM THE CHARM PHASE 3 TRIAL. In: Presented at the European Stroke Organisation Conference. 2024. Available at: https://eso-stroke.org/wp-content/uploads/Day_1_Clinical_Trial_Highlights_Full.pdf?_gl=1∗1n0mb97∗_up∗MQ.∗_ga∗MTcwMjY3MTA2MS4xNzIxMzc1NDg0∗_ga_DR13DEHDJ3∗MTcyMTM3NTQ4MS4xLjEuMTcyMTM3NTUxNy4wLjAuMA. [Accessed 19 July 2024]. [Google Scholar]
  • 69.Late break abstract - 2024. Eur Stroke J 2024; 9: (1 Suppl): 648–705.38353230 [Google Scholar]
  • 70.Zhao J, Song C, Li D, et al. GATE-ICH Study Group. Efficacy and safety of glibenclamide therapy after intracerebral haemorrhage (GATE-ICH): a multicentre, prospective, randomised, controlled, open-label, blinded-endpoint, phase 2 clinical trial. EClinicalMedicine 2022; 53:101666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.A multicenter, double-blind, multidose, placebo-controlled, randomized, parallel-group, phase 2 study to evaluate the efficacy and safety of intravenous BIIB093 for patients with brain contusion. Available at: https://www.clinicaltrials.gov/study/NCT03954041?tab=results. [Accessed 17 July 2024]. [Google Scholar]
  • 72.Wu T, Wu H, Wang J, et al. Expression and cellular localization of cyclooxygenases and prostaglandin E synthases in the hemorrhagic brain. J Neuroinflammation 2011; 8:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Chu K, Jeong SW, Jung KH, et al. Celecoxib induces functional recovery after intracerebral hemorrhage with reduction of brain edema and perihematomal cell death. J Cereb Blood Flow Metab 2004; 24:926–933. [DOI] [PubMed] [Google Scholar]
  • 74.Park HK, Lee SH, Chu K, Roh JK. Effects of celecoxib on volumes of hematoma and edema in patients with primary intracerebral hemorrhage. J Neurol Sci 2009; 279 (1–2):43–46. [DOI] [PubMed] [Google Scholar]
  • 75.Lee S-H, Park H-K, Ryu W-S, et al. Effects of celecoxib on hematoma and edema volumes in primary intracerebral hemorrhage: a multicenter randomized controlled trial. Eur J Neurol 2013; 20:1161–1169. [DOI] [PubMed] [Google Scholar]
  • 76.Chen CJ, Ding D, Ironside N, et al. Statins for neuroprotection in spontaneous intracerebral hemorrhage. Neurology 2019; 93:1056–1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Naval NS, Abdelhak TA, Urrunaga N, et al. An association of prior statin use with decreased perihematomal edema. Neurocrit Care 2008; 8:13–18. [DOI] [PubMed] [Google Scholar]
  • 78.Gioia LC, Kate M, McCourt R, et al. Perihematoma cerebral blood flow is unaffected by statin use in acute intracerebral hemorrhage patients. J Cereb Blood Flow Metab 2015; 35:1175–1180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Sprügel MI, Kuramatsu JB, Volbers B, et al. Impact of statins on hematoma, edema, seizures, vascular events, and functional recovery after intracerebral hemorrhage. Stroke 2021; 52:975–984. [DOI] [PubMed] [Google Scholar]
  • 80▪.Marchina S, Yeatts SD, Foster LD, et al. Rationale and design of the Statin Use in Intracerebral Hemorrhage Patients (SATURN) Trial. Cerebrovasc Dis 2024; [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]; This phase III trial aims to evaluate the impact of continuing versus discontinuing statins on ICH recurrence and cardiovascular events in lobar ICH survivors. It also investigates the role of APOE genotype in modifying these effects.
  • 81▪.Gao D, Chu X, Zhang Y, et al. Statins for neuroprotection in spontaneous intracerebral haemorrhage (STATIC): protocol for a multicentre, prospective and randomised controlled trial. BMJ Open 2024; 14:e079879. [DOI] [PMC free article] [PubMed] [Google Scholar]; This phase II/III trial aims to investigate the efficacy and safety of statins in reducing perihaemorrhagic oedema and improving outcomes in ICH patients, and provide evidence-based support for the use of statins in this context.
  • 82.Chojnowski K, Opiełka M, Gozdalski J, et al. The role of arginine-vasopressin in stroke and the potential use of arginine-vasopressin type 1 receptor antagonists in stroke therapy: a narrative review. Int J Mol Sci 2023; 24:2119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tan Q, Li Y, Guo P, et al. Tolvaptan attenuated brain edema in experimental intracerebral hemorrhage. Brain Res 2019; 1715:41–46. [DOI] [PubMed] [Google Scholar]
  • 84.Corry JJ, Asaithambi G, Shaik AM, et al. Conivaptan for the reduction of cerebral edema in intracerebral hemorrhage: a safety and tolerability study. Clin Drug Investig 2020; 40:503–509. [DOI] [PubMed] [Google Scholar]
  • 85.Der-Nigoghossian C, Lesch C, Berger K. Effectiveness and tolerability of conivaptan and tolvaptan for the treatment of hyponatremia in neurocritically ill patients. Pharmacotherapy 2017; 37:528–534. [DOI] [PubMed] [Google Scholar]
  • 86.Yankova G, Bogomyakova O, Tulupov A. The glymphatic system and meningeal lymphatics of the brain: new understanding of brain clearance. Rev Neurosci 2021; 32:693–705. [DOI] [PubMed] [Google Scholar]
  • 87.Iliff JJ, Wang M, Liao Y, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med 2012; 4:147ra111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Raposo N, Viswanathan A. MRI-visible enlarged perivascular spaces. Neurology 2020; 95:709–710. [DOI] [PubMed] [Google Scholar]
  • 89.Best JG, Barbato C, Ambler G, et al. Clinical Relevance of Microbleeds in Stroke (CROMIS-2) collaborators. Association of enlarged perivascular spaces and anticoagulant-related intracranial hemorrhage. Neurology 2020; 95.:e2192–e2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Liu X, Wu G, Tang N, et al. Glymphatic drainage blocking aggravates brain edema, neuroinflammation via modulating TNF-α, IL-10, and AQP4 after intracerebral hemorrhage in rats. Front Cell Neurosci 2021; 15:784154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Louveau A, Plog BA, Antila S, et al. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J Clin Invest 2017; 127:3210–3219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Tsai HH, Hsieh YC, Lin JS, et al. Functional investigation of meningeal lymphatic system in experimental intracerebral hemorrhage. Stroke 2022; 53:987–998. [DOI] [PubMed] [Google Scholar]

Articles from Current Opinion in Neurology are provided here courtesy of Wolters Kluwer Health

RESOURCES