Skip to main content

The intracranial compartmental syndrome: a proposed model for acute brain injury monitoring and management

Abstract

For decades, one of the main targets in the management of severe acute brain injury (ABI) has been intracranial hypertension (IH) control. However, the determination of IH has suffered variations in its thresholds over time without clear evidence for it. Meanwhile, progress in the understanding of intracranial content (brain, blood and cerebrospinal fluid) dynamics and recent development in monitoring techniques suggest that targeting intracranial compliance (ICC) could be a more reliable approach rather than guiding actions by predetermined intracranial pressure values. It is known that ICC impairment forecasts IH, as intracranial volume may rapidly increase inside the skull, a closed bony box with derisory expansibility. Therefore, an intracranial compartmental syndrome (ICCS) can occur with deleterious brain effects, precipitating a reduction in brain perfusion, thereby inducing brain ischemia. The present perspective review aims to discuss the ICCS concept and suggest an integrative model for the combination of modern invasive and noninvasive techniques for IH and ICC assessment. The theory and logic suggest that the combination of multiple ancillary methods may enhance ICC impairment prediction, pointing proactive actions and improving patient outcomes.

Introduction

For years, the evaluation and management of intracranial hypertension (IH), based on specific thresholds, have been the main target ("tip of the iceberg") for the treatment of acute brain injury (ABI), especially for traumatic brain injury (TBI) [1]. The increase in intracranial pressure (ICP) generates deleterious effects because of the displacement of anatomical structures, leading to a cascade of brain swelling, ischemia and generating different degrees and types of brain tissue herniation [2, 3].

Recently, an expert panel developed management algorithms for TBI care based on 22 mmHg for ICP threshold [4, 5]. Notwithstanding, such recommendations are supported only by lower evidence levels [6, 7]. In fact, the only randomized controlled trial for the management of TBI comparing ICP monitoring vs a clinical protocol guided by examination and neuroimaging (Best-Trip trial) demonstrated that ICP monitoring was not a necessary intervention when serial bedside neurological examination and brain imaging were taken [8]. This study changed the paradigm regarding the consideration of ICP as an isolated central intervention in TBI [9]. Moreover, it led to the emergence of arguments advocating against maintaining an empirical, fixed and rigid ICP cutoff as a pillar for starting different medical and/or surgical interventions [7, 10].

The secondary events after ABI are heterogeneous between subjects [11]. Moreover, even for the same patient, adjusting ideal cerebral perfusion may require arterial blood pressure (ABP) changes during periods of physiological instability following injury [11, 12]. Hence, determining the most vulnerable periods for tissue hypoxia and cellular dysfunction [13, 14] can be challenging [15,16,17,18]. In this context, intracranial compliance (ICC) impairment, the threshold with which intracranial volume has overpassed the inner compensatory reserve [19, 20], can be a more reliable target than ICP alone [21].

The recent advances in technology have brought intensive care units the opportunity to monitor closely and predict the undesired consequences of ICC impairment [22, 23], with a synergist diagnostic potential when these techniques are combined [24, 25]. Therefore, the modern management of IH should focus on a different and more integrated perspective, considering the instruments available to monitor these phenomena [26,27,28].

The present perspective review aims to propose the integration of monitoring techniques currently available to assess ICC impairment, such as ICP monitoring, transcranial Doppler (TCD), pupillometry, brain oximetry, near-infrared spectroscopy (NIRS), optic nerve sheath diameter ultrasound (ONSD) and automated ICP waveform analysis (ICPW). With these tools at hand, we propose a model and treatment algorithm utilizing the intracranial compartmental syndrome (ICCS) that may serve as an improvement in IH management. Mapping how different techniques can be associated is particularly important among locations where resources are scarce, such as low-income countries.

The intracranial compartmental syndrome

Definition

The ICCS is an ICC impairment diagnostic model applying different monitoring techniques with educational purpose as a standard of care. As for IH, ICCS occurs because the skull is a non-extensible compartment with limited adaptation to changes in pressure, and when the inner volume reaches a critical level, ICC is exhausted (Fig. 1) [20, 29,30,31]. The rigid cranial cavity is interconnected with other cavities as thorax and neck by venous and cerebrospinal fluid (CSF) systems, so IH may develop because of cranial and extracranial conditions (Table 1). As for any body cavity, when the inner pressure increases severely, it causes hypoperfusion, ischemia and tissue damage as a consequence of sensitive structures compression, such as nerves and blood vessels [32,33,34].

Fig. 1
figure 1

Different phases of the compensatory system. In the first phase (a), compensatory system is effective during a mass expansion. ICP does not change in this early phase, being ICC and the compensatory system adjusted. In a second phase (b), the compensatory system starts to fail following more increase in the mass effect. CSF and veins outflow are starting to be overloaded, beginning brain deformation and ICC impairment. In a third phase (c), the compensatory system is completely exhausted, and brain deformation and loss of ICC are evident. ICP: intracranial pressure, ICC: intracranial compliance, CSF: cerebrospinal fluid. Adapted from Wykes et al. [31]

Table 1 Causes of intracranial hypertension

In early stages of ICCS development, a space-occupying lesion (contusion) or an increase in brain parenchyma volume (edema) does not cause an increase in ICP, so long as the compensation systems and cerebral autoregulation work [3]. If the process is not aborted at this time, ICP will increase exponentially, compromising perfusion, oxygenation, energy usage and creating compartmental gradients that will anatomically distorting brain tissue. It is important to remark that these changes are not necessarily associated with specific ICP number thresholds, as we can find patients with loss of ICC within a predetermined “normal range” of ICP or in patients with preserved ICC that demonstrate ICP above these thresholds (Fig. 2).

Fig. 2
figure 2

Proposed integrative model. Stage 1: normal ICC, stage 2: ICC impairment and stage 3: severe ICC impairment. Types A-D explained in detail in the text. ICP: intracranial pressure. ICC: intracranial compliance, ICPW: ICP waveform, NIRS: near-infrared spectroscopy, PtiO2: cerebral oximetry

Therefore, the hallmark of reduction in ICC must rely on the ICP pulse morphology or waveform (ICPW) [35]. ICPW has been extensively studied and constitutes the leading monitor in the ICCS diagnostic toolbox. The changes in ICP pulse morphology have been directly linked to ICC impairment, especially when the second peak (P2) assumes a higher amplitude than the first peak (P1, Fig. 3), forecasting IH [36, 37]. In combination with changes in ICPW, other invasive and noninvasive methods can be added as synergistic adjuncts to monitor brain oxygenation, compliance and blood dynamics.

Fig. 3
figure 3

ICP waves registered at 25 mm per second showing the three components (P1, P2 and P3). a Normal pattern; b pattern of impaired compliance. ICP: intracranial pressure (Source: authors)

Thresholds

ICP probes may display a spectrum of measured values, from exclusively ICP mean values up to ICP trends, systolic and diastolic values, brain temperature and waveforms [38]. Without an automated analysis of ICPW from invasive methods, the contour analysis relies on subjectivity and expertise to conclude when P2 surpasses P1 in amplitude [39]. Oximetry probes may also provide brain temperature and a local (around 2.5 cc) brain tissue oxygenation (PbtO2), with ideal around 20–35 mmHg [40], whereas NIRS and jugular venous oxygen saturation (SvjO2) provide percentages of hemoglobin oxygen saturation. For ICCS, we used ICP > 20 mmHg and PbtO2 < 20 mmHg [41] or SO2 < 50% if NIRS or SvjO2 are used [42].

An ICU validated, noninvasive and mobile technique to monitor ICP variations based on ICPW has been recently entered the market (Braincare Corp, São Carlos, Brazil) [38, 43,44,45,46]. The system is based on cranial micrometric deformation; it currently does not provide ICP values, but extracts parametric values from the pulse slopes that are correlated with IH [38]. For ICCS, we applied the P2/P1 ratio > 1.2 provided by this technique as indicator of IH [44].

Ultrasound techniques, such as duplex and TCD, can be useful in several neurovascular diagnostic areas: cerebral autoregulation assessment [3], embolic activity [47], arterial [48] and venous stenosis, as well as supportive evidence in brain death [49]. TCD acquires waveforms derived from blood velocities and may indicate reduction in cerebral perfusion pressure through dedicated software [22], the pulsatility index (PI) and/or reduction in mean velocities [25]. Duplex also can observe intracranial hematomas and middle line shift [50], and evaluate the ONSD. It has also demonstrated excellent negative predictive value for the estimation of ICP [51]. For ICCS, we used the PI > 1.2 and ONSD > 5 mm as indicators of IH [52].

Pupillometry can provide sedation status, assessment of pain, prediction of clinical deterioration and outcome [53]. Although it has reduced capacity to detect IH by the pupillary reflex alone, the automated neurological pupil index (NPi) present in dedicated manufacturer (NeurOptics, Irvine, USA) is reliable to observe worsening in neurological condition as consequence of IH, when serial measures are performed [53]. For ICCS, we used the NPi < 3 as an indicator of neurological deterioration. Main techniques advantages and limitations included in the model are summarized in Table 2.

Table 2 Characteristics of most relevant noninvasive surrogate techniques for ICP monitoring

EEG has not been included, but should be considered as an additional information to this ICCS algorithm. Furthermore, metabolic crisis [54] and spreading depolarizations [55] are examples of real menaces for ABI patients that can occur unnoticed in this model [56], being a limitation of ICCS.

Proposed diagnostic model

The proposed model integrates the monitoring of ICC through the analysis of ICPW to the traditional invasive and/or noninvasive monitoring methods of ICP and cerebral tissue oxygenation (Fig. 4). The techniques included have a solid and compelling rationale for their use, despite the fact that large trials remain lacking [57, 58]. As many of these techniques are referred to physiological phenomena, it is not expected that all techniques show their results exactly as presented below, but inconclusive results may guide continuous/serial monitoring and revisiting the patient records. Considering the variety of resources from one location to another, as well as the advantages and limitations of each technique, this is not a condition to be assessed exclusively if all methods are available. Rather, it is a recommendation for practitioners to take hand of all resources present; the more the information available, the more likely an assertive decision will result. Of course, the comprehensive management of ICCS depends on a thorough review of the medical record and available brain imaging.

Fig. 4
figure 4

Integrative model of multimodal monitoring with the thresholds for different invasive and noninvasive techniques. Types A-D explained in detail in the text. ICP: intracranial pressure. ICPW: ICP waveform, NIRS: near-infrared spectroscopy, PtiO2: cerebral oximetry

Four evolutionary patterns are proposed, utilizing the ICPW characterization:

Type A: normal pattern. Absence of intracranial hypertension and tissue hypoxia on invasive and noninvasive monitoring, i.e., preserved ICPW.

Type B: IH without impairment of cerebral oxygenation while maintaining ICC (borderline ICPW). Such a pattern may be seen in insidious chronic conditions such as obesity [59], in the early stages of hydrocephalus development [60] or during the early stages of space-occupying lesions or cerebral edema [61]. Furthermore, extracranial causes of increased ICP (pneumothorax, mechanical ventilator asynchrony [62, 63], airway obstruction, intrabdominal hypertension [64]).

Type C: Grade I ICCS. Alteration of ICC evidenced through the change in the ICP pulse morphology (P2 > or = P1), in the “absence of an increase in the numerical value of the ICP.” Causes of this pattern are temporal or frontal contusions < 25 cc (diffuse injury type II of the Marshall’s tomographic classification) or laminar extra-axial lesions that do not cause midline deviation. Depending on the evolutionary stage, it may or may not be accompanied (advanced) by brain tissue hypoxia (early).

Type D: Grade II ICCS. The syndrome in its fullness, as a life-threatening condition. It is characterized by total loss of compliance with the presence of marked morphological changes in the ICPW with effacement of the P1 component and adoption of a pyramidal shape, accompanied by IH and cerebral tissue hypoxia. Pattern that can be observed in non-evacuated space-occupying lesions > 25 cc or diffuse type III or IV injury of the Marshall’s classification.

Therapeutical approach

Following the pathophysiological reasoning of the proposed model, the therapeutic approach can be based on the following premises:

Type A: Treatment will be based on clinical, imaging, hemodynamic, metabolic and intracranial oxygenation monitoring. From their analysis, the intensivist will proceed to the implementation of general care measures, which may include physiological neuroprotection, mechanical ventilation and sedation/analgesia, avoiding secondary insults, and preventing deep vein thrombosis, gastrointestinal bleeding and infectious complication [65]. Anticonvulsants when indicated, early nutrition and rehabilitation are important additional measures. This global approach should be continuous even for the other subtypes [4, 6, 65, 66].

Type B: IH without ICC impairment. Before starting therapy, it is important to carry out an exhaustive analysis of the cause (whether intra- or extracranial, for example), since the therapy will depend on etiology [66]. In case of hydrocephalus, external ventricular drainage will be the choice; while if the origin is increased intrathoracic pressure, due to asynchrony with mechanical ventilation, deepening sedation/analgesia after analysis of the ventilatory mode will be priorities.

Type C: ICCS grade I. IH may or may not be present, but ICC impairment leads to brain tissue oxygenation alteration. This situation is probably the most difficult to defining therapeutics. Although initial medical management of IH and cerebral tissue hypoxia is based on individual institutional guidelines or international consensus [4, 66, 67], it is essential to keep in mind the following premises: (a) close, continued follow-up monitoring and further therapeutic response based on wave morphology [68, 69]; (b) refrain from escalation of any medical treatment beyond recommended levels, if the response to that intervention is not satisfactory; and (c) early consideration of CSF drainage and/or surgical evacuation of space-occupying injuries or decompression of the cranial cavity.

Type D: ICCS grade II. The syndrome is fully developed. Combined medical and surgical therapy is mandatory, but prompt consideration of the latter is considered essential, since decompression of the cranial cavity is urgently necessary independent of the nature (whether focal or diffuse) of the lesions [70, 71].

Conclusions and future perspectives

Modern management of ABI has broken the simplistic intracranial hypertension-based model of care. Other phenomena such as brain tissue hypoxia and energy dysfunction are important to recognize, prevent and treat to optimize results. Severe TBI is dynamic and heterogeneous. The advancement and analysis of multimodal monitoring brought with it the concept of “personalized therapy.” The proposed model integrates the monitoring of intracranial compliance with the traditional monitored variables (invasive or noninvasive) during severe TBI. The ICCS is defined not by a specific numeric threshold of ICP monitoring, but based on its subtype and multimodal monitoring, suggesting therapeutic approaches for emergency conditions. Large-scale studies are necessary to evaluate this proposed model in addition to validate the use of new noninvasive monitoring techniques for understanding these new concepts. We advocate for moving toward new concepts and paradigm shifts in the management of ABI in order to decrease mortality and disability associated with a delayed decision-making process.

Availability of data and materials

Not applicable.

Abbreviations

TBI:

Traumatic brain injury

ICP:

Intracranial pressure

ICPW:

Intracranial pressure waveform

CSF:

Cerebrospinal fluid

MLS:

Midline shift

CT:

Computed tomography

IH:

Intracranial hypertension

ICCS:

Intracranial compartmental syndrome

CBV:

Cerebral blood volume

ICC:

Intracranial compliance

CPP:

Cerebral perfusion pressure

MAP:

Mean arterial pressure

TCD:

Transcranial Doppler

NPV:

Negative predictive value

B4C:

Brain4care

TTP:

Time to peak

ABI:

Acute brain injury

References

  1. Rabelo NN, da Silva BJ, da Silva JS, de Souza NB, Coelho G, Brasil S, Frigieri G. The historic evolution of intracranial pressure and cerebrospinal fluid pulse pressure concepts: Two centuries of challenges. Surg Neurol Int. 2021;12:274.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Brain Trauma F, American Association of Neurological S, Congress of Neurological S, Joint Section on N, Critical Care AC, Bratton SL, Chestnut RM, Ghajar J, McConnell Hammond FF, Harris OA et al. Guidelines for the management of severe traumatic brain injury. VIII. Intracranial pressure thresholds. J Neurotrauma. 2007;24 Suppl 1:S55–58.

  3. Brasil S, Nogueira RC, Salinet ASM, Yoshikawa MH, Teixeira MJ, Paiva W, Malbouisson LMS, Bor-Seng-Shu E, Panerai RB. The contribution of intracranial pressure to human dynamic cerebral autoregulation after acute brain injury. Am J Physiol Regul Integr Comp Physiol. 2022.

  4. Chesnut R, Aguilera S, Buki A, Bulger E, Citerio G, Cooper DJ, Arrastia RD, Diringer M, Figaji A, Gao G, et al. A management algorithm for adult patients with both brain oxygen and intracranial pressure monitoring: the Seattle International Severe Traumatic Brain Injury Consensus Conference (SIBICC). Intensive Care Med. 2020;46(5):919–29.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Grande PO. The “Lund Concept” for the treatment of severe head trauma—physiological principles and clinical application. Intensive Care Med. 2007;33(1):205.

    Article  PubMed  Google Scholar 

  6. Carney N, Totten AM, O’Reilly C, Ullman JS, Hawryluk GW, Bell MJ, Bratton SL, Chesnut R, Harris OA, Kissoon N, et al. Guidelines for the management of severe traumatic brain injury, fourth edition. Neurosurgery. 2017;80(1):6–15.

    Article  PubMed  Google Scholar 

  7. Wijdicks EFM. 10 or 15 or 20 or 40 mmHg? What is Increased intracranial pressure and who said so? Neurocrit Care. 2022;36(3):1022–6.

    Article  PubMed  Google Scholar 

  8. Chesnut RM, Temkin N, Carney N, Dikmen S, Rondina C, Videtta W, Petroni G, Lujan S, Pridgeon J, Barber J, et al. A trial of intracranial-pressure monitoring in traumatic brain injury. N Engl J Med. 2012;367(26):2471–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lazaridis C, Desai M, Damoulakis G, Zeiler FA. Intracranial pressure threshold heuristics in traumatic brain injury: one, none, many! Neurocrit Care. 2020;32(3):672–6.

    Article  PubMed  Google Scholar 

  10. Chesnut RM, Videtta W. Situational intracranial pressure management: an argument against a fixed treatment threshold. Crit Care Med. 2020;48(8):1214–6.

    Article  PubMed  Google Scholar 

  11. Stocchetti N, Carbonara M, Citerio G, Ercole A, Skrifvars MB, Smielewski P, Zoerle T, Menon DK. Severe traumatic brain injury: targeted management in the intensive care unit. Lancet Neurol. 2017;16(6):452–64.

    Article  PubMed  Google Scholar 

  12. Nourallah B, Zeiler FA, Calviello L, Smielewski P, Czosnyka M, Menon DK. Critical thresholds for intracranial pressure vary over time in non-craniectomised traumatic brain injury patients. Acta Neurochir (Wien). 2018;160(7):1315–24.

    Article  PubMed  Google Scholar 

  13. Brasil S, Paiva WS, de Carvalho Nogueira R, Macedo Salinet A, Teixeira MJ. Letter to the Editor. Decompressive craniectomy in TBI: What is beyond static evaluations in terms of prognosis? J Neurosurg. 2018;129(3):845–47.

  14. Bouzat P, Sala N, Payen JF, Oddo M. Beyond intracranial pressure: optimization of cerebral blood flow, oxygen, and substrate delivery after traumatic brain injury. Ann Intensive Care. 2013;3(1):23.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Vik A, Nag T, Fredriksli OA, Skandsen T, Moen KG, Schirmer-Mikalsen K, Manley GT. Relationship of “dose” of intracranial hypertension to outcome in severe traumatic brain injury. J Neurosurg. 2008;109(4):678–84.

    Article  PubMed  Google Scholar 

  16. Guiza F, Depreitere B, Piper I, Citerio G, Chambers I, Jones PA, Lo TY, Enblad P, Nillson P, Feyen B, et al. Visualizing the pressure and time burden of intracranial hypertension in adult and paediatric traumatic brain injury. Intensive Care Med. 2015;41(6):1067–76.

    Article  PubMed  Google Scholar 

  17. Jha RM, Elmer J, Zusman BE, Desai S, Puccio AM, Okonkwo DO, Park SY, Shutter LA, Wallisch JS, Conley YP, et al. Intracranial pressure trajectories: a novel approach to informing severe traumatic brain injury phenotypes. Crit Care Med. 2018;46(11):1792–802.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Yang F, Peng C, Peng L, Wang P, Cheng C, Zuo W, Zhao L, Jin Z, Li W. Group-based trajectory modeling of intracranial pressure in patients with acute brain injury: results from multi-center ICUs, 2008–2019. CNS Neurosci Ther. 2022;28(8):1218–28.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Zeiler FA, Kim DJ, Cabeleira M, Calviello L, Smielewski P, Czosnyka M. Impaired cerebral compensatory reserve is associated with admission imaging characteristics of diffuse insult in traumatic brain injury. Acta Neurochir (Wien). 2018;160(12):2277–87.

    Article  PubMed  Google Scholar 

  20. Rubiano AM, Figaji A, Hawryluk GW. Intracranial pressure management: moving beyond guidelines. Curr Opin Crit Care. 2022;28(2):101–10.

    Article  PubMed  Google Scholar 

  21. Ocamoto GN, Russo TL, Mendes Zambetta R, Frigieri G, Hayashi CY, Brasil S, Rabelo NN, Spavieri Junior DL. Intracranial compliance concepts and assessment: a scoping review. Front Neurol. 2021;12: 756112.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Brasil S, de Carvalho Nogueira R, Salinet  SM, Yoshikawa MH, Teixeira MJ, Paiva W, Malbouisson LMS, Bor-Seng-Shu E, Panerai RB. Critical closing pressure and cerebrovascular resistance responses to intracranial pressure variations in neurocritical patients. Neurocrit Care. 2023.

  23. Hawryluk GWJ, Citerio G, Hutchinson P, Kolias A, Meyfroidt G, Robba C, Stocchetti N, Chesnut R. Intracranial pressure: current perspectives on physiology and monitoring. Intensive Care Med. 2022.

  24. Robba C, Frigieri G, Brasil S, Taccone FS. Early prognostic value of non-invasive intracranial pressure methods in brain-injured patients. Intensive Care Med. 2022.

  25. Robba C, Pozzebon S, Moro B, Vincent JL, Creteur J, Taccone FS. Multimodal non-invasive assessment of intracranial hypertension: an observational study. Crit Care. 2020;24(1):379.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Lauerman MH, Stein DM. Multicompartment management of patients with severe traumatic brain injury. Curr Opin Anaesthesiol. 2014;27(2):219–24.

    Article  PubMed  Google Scholar 

  27. Scalea TM, Bochicchio GV, Habashi N, McCunn M, Shih D, McQuillan K, Aarabi B. Increased intra-abdominal, intrathoracic, and intracranial pressure after severe brain injury: multiple compartment syndrome. J Trauma. 2007;62(3):647–656; discussion 656.

  28. Wilson MH. Monro-Kellie 2.0: the dynamic vascular and venous pathophysiological components of intracranial pressure. J Cereb Blood Flow Metab. 2016;36(8):1338–1350.

  29. Brogi E, Coccolini F, Russo E, Forfori F. Diagnosis and treatment of the intracranial compartment syndrome. In: Coccolini F, Malbrain MLNG, Kirkpatrick AW, Gamberini E, editors. Compartment syndrome. Cham: Springer; 2021. p. 17–33.

    Chapter  Google Scholar 

  30. Figaji AA, Fieggen AG, Argent A, Peter JC. Surgical treatment for “brain compartment syndrome” in children with severe head injury. S Afr Med J. 2006;96(9 Pt 2):969–75.

    CAS  PubMed  Google Scholar 

  31. Wykes V, Vindlacheruvu R. Intracranial pressure, cerebral blood flow and brain oedema. Surg Infect (Larchmt). 2015;33(8):355–62.

    Google Scholar 

  32. Gourgiotis S, Villias C, Germanos S, Foukas A, Ridolfini MP. Acute limb compartment syndrome: a review. J Surg Educ. 2007;64(3):178–86.

    Article  PubMed  Google Scholar 

  33. Pereira BM. Abdominal compartment syndrome and intra-abdominal hypertension. Curr Opin Crit Care. 2019;25(6):688–96.

    Article  PubMed  Google Scholar 

  34. Pereira R, Buglevski M, Perdigoto R, Marcelino P, Saliba F, Blot S, Starkopf J. Intra-abdominal hypertension and abdominal compartment syndrome in the critically ill liver cirrhotic patient-prevalence and clinical outcomes. A multicentric retrospective cohort study in intensive care. PLoS ONE. 2021;16(5):e0251498.

  35. Brasil S. Intracranial pressure pulse morphology: the missing link? Intensive Care Med. 2022.

  36. Hamilton R, Xu P, Asgari S, Kasprowicz M, Vespa P, Bergsneider M, Hu X. Forecasting intracranial pressure elevation using pulse waveform morphology. Annu Int Conf IEEE Eng Med Biol Soc. 2009;2009:4331–4.

    PubMed  Google Scholar 

  37. Hu X, Xu P, Asgari S, Vespa P, Bergsneider M. Forecasting ICP elevation based on prescient changes of intracranial pressure waveform morphology. IEEE Trans Biomed Eng. 2010;57(5):1070–8.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Frigieri G, Robba C, Machado FS, Gomes JA, Brasil S. Application of non-invasive ICP waveform analysis in acute brain injury: Intracranial Compliance Scale. Intensive Care Med Exp. 2023;11(1):5.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Czosnyka M, Czosnyka Z. Origin of intracranial pressure pulse waveform. Acta Neurochir (Wien). 2020;162(8):1815–7.

    Article  PubMed  Google Scholar 

  40. Maloney-Wilensky E, Le Roux P. The physiology behind direct brain oxygen monitors and practical aspects of their use. Childs Nerv Syst. 2010;26(4):419–30.

    Article  PubMed  Google Scholar 

  41. Battaglini D, Anania P, Rocco PRM, Brunetti I, Prior A, Zona G, Pelosi P, Fiaschi P. Escalate and De-escalate therapies for intracranial pressure control in traumatic brain injury. Front Neurol. 2020;11: 564751.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Godoy DA, Murillo-Cabezas F, Suarez JI, Badenes R, Pelosi P, Robba C. “THE MANTLE” bundle for minimizing cerebral hypoxia in severe traumatic brain injury. Crit Care. 2023;27(1):13.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Brasil S, Frigieri G, Taccone FS, Robba C, Solla DJF, de Carvalho Nogueira R, Yoshikawa MH, Teixeira MJ, Malbouisson LMS, Paiva WS. Noninvasive intracranial pressure waveforms for estimation of intracranial hypertension and outcome prediction in acute brain-injured patients. J Clin Monit Comput. 2022.

  44. Brasil S, Solla DJF, Nogueira RC, Teixeira MJ, Malbouisson LMS, Paiva WS. A novel noninvasive technique for intracranial pressure waveform monitoring in critical care. J Pers Med. 2021;11(12):1302.

    Article  PubMed  PubMed Central  Google Scholar 

  45. de Moraes FM, Rocha E, Barros FCD, Freitas FGR, Miranda M, Valiente RA, de Andrade JBC, Neto F, Silva GS. Waveform morphology as a surrogate for ICP monitoring: a comparison between an invasive and a noninvasive method. Neurocrit Care. 2022.

  46. Hassett CE, Uysal SP, Butler R, Moore NZ, Cardim D, Gomes JA. Assessment of cerebral autoregulation using invasive and noninvasive methods of intracranial pressure monitoring. Neurocrit Care. 2022.

  47. Paschoal FM Jr, de Almeida Lins Ronconi K, de Lima Oliveira M, Nogueira RC, Paschoal EH, Teixeira MJ, Figueiredo EG, Bor-Seng-Shu E. Embolic signals during routine transcranial Doppler ultrasonography in aneurysmal subarachnoid hemorrhage. Biomed Res Int. 2015;2015:153714.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Bor-Seng-Shu E, de Lima-Oliveira M, Teixeira MJ, Panerai RB. Predicting symptomatic cerebral vasospasm after aneurysmal subarachnoid hemorrhage. Neurosurgery. 2011;69(2):E501-502.

    Article  PubMed  Google Scholar 

  49. Brasil S, Bor-Seng-Shu E, de-Lima-Oliveira M, Taccone FS, Gattas G, Nunes DM, Gomes de Oliveira RA, Martins Tomazini B, Tierno PF, Becker RA et al. Computed tomography angiography accuracy in brain death diagnosis. J Neurosurg. 2019:1–9.

  50. Vitiello L, Salerno G, De Bernardo M, D’Aniello O, Capasso L, Marotta G, Rosa N. Ultrasound detection of intracranial hypertension in brain injuries. Front Med (Lausanne). 2022;9: 870808.

    Article  PubMed  Google Scholar 

  51. Rasulo FA, Calza S, Robba C, Taccone FS, Biasucci DG, Badenes R, Piva S, Savo D, Citerio G, Dibu JR, et al. Transcranial Doppler as a screening test to exclude intracranial hypertension in brain-injured patients: the IMPRESSIT-2 prospective multicenter international study. Crit Care. 2022;26(1):110.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Rasulo FA, Bertuetti R. Transcranial doppler and optic nerve sonography. J Cardiothorac Vasc Anesth. 2019;33(Suppl 1):S38–52.

    Article  PubMed  Google Scholar 

  53. Sandroni C, Citerio G, Taccone FS. Automated pupillometry in intensive care. Intensive Care Med. 2022;48(10):1467–70.

    Article  PubMed  Google Scholar 

  54. de Lima OM, Kairalla AC, Fonoff ET, Martinez RC, Teixeira MJ, Bor-Seng-Shu E. Cerebral microdialysis in traumatic brain injury and subarachnoid hemorrhage: state of the art. Neurocrit Care. 2014;21(1):152–62.

    Google Scholar 

  55. Hinzman JM, Wilson JA, Mazzeo AT, Bullock MR, Hartings JA. Excitotoxicity and metabolic crisis are associated with spreading depolarizations in severe traumatic brain injury patients. J Neurotrauma. 2016;33(19):1775–83.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Vespa P, Bergsneider M, Hattori N, Wu HM, Huang SC, Martin NA, Glenn TC, McArthur DL, Hovda DA. Metabolic crisis without brain ischemia is common after traumatic brain injury: a combined microdialysis and positron emission tomography study. J Cereb Blood Flow Metab. 2005;25(6):763–74.

    Article  CAS  PubMed  Google Scholar 

  57. Bernard F, Barsan W, Diaz-Arrastia R, Merck LH, Yeatts S, Shutter LA. Brain Oxygen Optimization in Severe Traumatic Brain Injury (BOOST-3): a multicentre, randomised, blinded-endpoint, comparative effectiveness study of brain tissue oxygen and intracranial pressure monitoring versus intracranial pressure alone. BMJ Open. 2022;12(3): e060188.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Citerio G, Oddo M, Taccone FS. Recommendations for the use of multimodal monitoring in the neurointensive care unit. Curr Opin Crit Care. 2015;21(2):113–9.

    Article  PubMed  Google Scholar 

  59. Subramaniam S, Fletcher WA. Obesity and weight loss in idiopathic intracranial hypertension: a narrative review. J Neuroophthalmol. 2017;37(2):197–205.

    Article  PubMed  Google Scholar 

  60. Schmidt EA, Despas F, Pavy-Le Traon A, Czosnyka Z, Pickard JD, Rahmouni K, Pathak A, Senard JM. Intracranial pressure is a determinant of sympathetic activity. Front Physiol. 2018;9:11.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Koenig MA. Cerebral edema and elevated intracranial pressure. Continuum (Minneap Minn). 2018;24(6):1588–602.

    PubMed  Google Scholar 

  62. Videtta W, Villarejo F, Cohen M, Domeniconi G, Santa Cruz R, Pinillos O, Rios F, Maskin B. Effects of positive end-expiratory pressure on intracranial pressure and cerebral perfusion pressure. Acta Neurochir Suppl. 2002;81:93–7.

    CAS  PubMed  Google Scholar 

  63. Robba C, Ball L, Nogas S, Battaglini D, Messina A, Brunetti I, Minetti G, Castellan L, Rocco PRM, Pelosi P. Effects of positive end-expiratory pressure on lung recruitment, respiratory mechanics, and intracranial pressure in mechanically ventilated brain-injured patients. Front Physiol. 2021;12: 711273.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Bloomfield GL, Ridings PC, Blocher CR, Marmarou A, Sugerman HJ. A proposed relationship between increased intra-abdominal, intrathoracic, and intracranial pressure. Crit Care Med. 1997;25(3):496–503.

    Article  CAS  PubMed  Google Scholar 

  65. Godoy DA, Videtta W, Santa Cruz R, Silva X, Aguilera-Rodriguez S, Carreno-Rodriguez JN, Ciccioli F, Pinero G, Ciro JD, da Re-Gutierrez S, et al. General care in the management of severe traumatic brain injury: Latin American consensus. Med Intensiva (Engl Ed). 2020;44(8):500–8.

    Article  CAS  PubMed  Google Scholar 

  66. Godoy DA, Videtta W, Di Napoli M. Practical approach to posttraumatic intracranial hypertension according to pathophysiologic reasoning. Neurol Clin. 2017;35(4):613–40.

    Article  PubMed  Google Scholar 

  67. Godoy DA, Lubillo S, Rabinstein AA. Pathophysiology and management of intracranial hypertension and tissular brain hypoxia after severe traumatic brain injury: an integrative approach. Neurosurg Clin N Am. 2018;29(2):195–212.

    Article  PubMed  Google Scholar 

  68. Liu X, Vitt JR, Hetts SW, Gudelunas K, Ho N, Ko N, Hu X. Morphological changes of intracranial pressure quantifies vasodilatory effect of verapamil to treat cerebral vasospasm. J Neurointerv Surg. 2020;12(8):802–8.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Huser M, Kundig A, Karlen W, De Luca V, Jaggi M. Forecasting intracranial hypertension using multi-scale waveform metrics. Physiol Meas. 2020;41(1): 014001.

    Article  PubMed  Google Scholar 

  70. Charry JD, Rubiano AM, Puyana JC, Carney N, David Adelson P. Damage control of civilian penetrating brain injuries in environments of low neuro-monitoring resources. Br J Neurosurg. 2016;30(2):235–9.

    Article  PubMed  Google Scholar 

  71. Rubiano AM, Maldonado M, Montenegro J, Restrepo CM, Khan AA, Monteiro R, Faleiro RM, Carreno JN, Amorim R, Paiva W, et al. The evolving concept of damage control in neurotrauma: application of military protocols in civilian settings with limited resources. World Neurosurg. 2019;125:e82–93.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Renato Abe for the illustration artwork.

Funding

The authors of this manuscript declare they have not received funds or fees for writing it.

Author information

Authors and Affiliations

Authors

Contributions

DAG proposed the original idea, wrote the first draft, produced original tables and figures, and discussed and edited the final version. AR proposed the original idea, and took part in the discussion, critical analysis and edition of the manuscript. SB drafted, revised and edited the whole manuscript and produced final tables and figures. WP contributed to illustrative case and discussion, carried out the critical analysis and edited the manuscript. CI participated in discussion, critical analysis and edition of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Sérgio Brasil.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

SB is consultant for Brain4care. The other authors declare no competing interests.

Additional information

Publisher's Note

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

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Godoy, D.A., Brasil, S., Iaccarino, C. et al. The intracranial compartmental syndrome: a proposed model for acute brain injury monitoring and management. Crit Care 27, 137 (2023). https://doi.org/10.1186/s13054-023-04427-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13054-023-04427-4

Keywords