Next Article in Journal
Thrombocytopenia among Patients Undergoing Aortic Valve Replacement Using the Sutureless Perceval S Bioprosthesis: A Retrospective Study
Next Article in Special Issue
Association between Unplanned Conversion and Patient Survival after Laparoscopic Liver Resection for Hepatocellular Carcinoma: A Propensity Score Matched Analysis
Previous Article in Journal
Unmet Horizons: Assessing the Challenges in the Treatment of TP53-Mutated Acute Myeloid Leukemia
Previous Article in Special Issue
Intrathecal Morphine and Post-Operative Pain Relief in Robotic Surgeries: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Underneath Images and Robots, Looking Deeper into the Pneumoperitoneum: A Narrative Review

by
Guido Mazzinari
1,2,3,*,
Lucas Rovira
4,
Kim I. Albers-Warlé
5,6,
Michiel C. Warlé
7,
Pilar Argente-Navarro
1,
Blas Flor
4 and
Oscar Diaz-Cambronero
1,2
1
Perioperative Medicine Research Group, Health Research Institute la Fe, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain
2
Department of Anesthesiology, La Fe University Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain
3
Department of Statistics and Operational Research, University of Valencia, Calle Doctor Moliner 50, 46100 Burjassot, Spain
4
Department of Anesthesiology, Consorcio Hospital General Universitario de Valencia, Av. de les Tres Creus, 2, L’Olivereta, 46014 València, Spain
5
Department of Colorectal Surgery, La Fe University Hospital, Avenida Fernando Abril Martorell 106, 46026 Valencia, Spain
6
Department of Anesthesiology, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
7
Departments of Surgery, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(4), 1080; https://doi.org/10.3390/jcm13041080
Submission received: 18 January 2024 / Revised: 5 February 2024 / Accepted: 10 February 2024 / Published: 14 February 2024
(This article belongs to the Special Issue New Insights in Laparoscopic Surgery)

Abstract

:
Laparoscopy offers numerous advantages over open procedures, minimizing trauma, reducing pain, accelerating recovery, and shortening hospital stays. Despite other technical advancements, pneumoperitoneum insufflation has received little attention, barely evolving since its inception. We explore the impact of pneumoperitoneum on patient outcomes and advocate for a minimally invasive approach that prioritizes peritoneal homeostasis. The nonlinear relationship between intra-abdominal pressure (IAP) and intra-abdominal volume (IAV) is discussed, emphasizing IAP titration to balance physiological effects and surgical workspace. Maintaining IAP below 10 mmHg is generally recommended, but factors such as patient positioning and surgical complexity must be considered. The depth of neuromuscular blockade (NMB) is explored as another variable affecting laparoscopic conditions. While deep NMB appears favorable for surgical stillness, achieving a balance between IAP and NMB depth is crucial. Temperature and humidity management during pneumoperitoneum are crucial for patient safety and optical field quality. Despite the debate over the significance of temperature drop, humidification and the warming of insufflated gas offer benefits in peritoneal homeostasis and visual clarity. In conclusion, there is potential for a paradigm shift in pneumoperitoneum management, with dynamic IAP adjustments and careful control of insufflated gas temperature and humidity to preserve peritoneal homeostasis and improve patient outcomes in minimally invasive surgery.

1. Towards the Minimally Invasive Pneumoperitoneum in Laparoscopy

Laparoscopic access has improved surgical results compared to open surgery, and it is now regarded as the gold standard for many indications [1,2,3,4,5]. It induces less direct trauma with smaller incisions. It has been associated with less postoperative pain, less systemic immunological depression, less wound infection, fewer complications, faster bowel recovery, shorter hospital stays, and earlier return to normal activities compared to open surgery [6]. Since its inception [7], apart from the continuous evolution of surgical instruments [8], two major advancements have been implemented. The first is related to image quality enhancement introduced by high definition and 3D depth [9,10], and the second is linked to the accuracy offered by robotic surgical procedures [11,12]. All these technical aids are aimed at facilitating the work of the surgical team and indirectly benefit the patient [13].
Conversely, the insufflation process, another critical component of the laparoscopy technique, has experienced minimal modifications. Its principle is to expand the virtual cavity that resides between the peritoneal membrane layers. However, the peritoneal membrane is far from inert. Its mesothelial cells are flattened squamous cells with epithelial-like microvilli that produce various substances like glycosaminoglycans, phospholipids, proteoglycans, surfactants, and coagulant precursors, forming a protective glycocalyx barrier. This barrier prevents shear stress, infections, and cancer spread, allowing the smooth sliding of serosal surfaces. Moreover, mesothelial cells are connected by tight junctions, integrins, and cadherins, anchored to the actin cytoskeleton, and produce fibronectin, laminin, and collagen, forming the basal membrane. Ultimately, they control fluid and cell transport, inflammation, tissue repair, fibrin deposit lysis, protection against microorganisms, and, potentially, tumor dissemination in the coelomic cavity [14]. Therefore, while gas insufflation in the abdominal cavity, i.e., the pneumoperitoneum, generates a suitable workspace for the surgeon, it can potentially impair the patient’s peritoneal homeostasis by causing reduced peritoneal blood flow, peritoneal acidosis, oxidative stress, mesothelial alterations, and the formation of adhesions [15]. The pneumoperitoneum’s chemical, physical, and biological features may affect clinical outcomes such as postoperative pain, recovery time, and the length of hospital stay [16,17,18]. The injury that occurs with conventional insufflation is due to the variation in three fundamental parameters: the gas psychometric features (i.e., temperature and humidity) and the amount of pressure and gas volume administered. In this narrative review, we will review the mechanism of injury and which implementations can potentially reduce it.

2. Pneumoperitoneum Effects and the Pressure–Volume Derivative

Even though the European Association for Endoscopic Surgery suggests adjusting intra-abdominal pressure (IAP) to the minimum level necessary for a sufficient surgical workspace, it is a widespread clinical practice to maintain a constant IAP between 12 and 15 mmHg [19]. This approach is based on the dubious assumption that applying more pressure always allows for more space. Yet, like all bodies with rigid borders, the abdominal cavity cannot expand indefinitely. According to biomechanics laws, materials initially undergo an elastic phase where there is almost a linear relationship between the stress, i.e., the force applied, and strain, i.e., the deformation that they experience. Eventually, a point of maximum stretch (the yield stress) is reached, where additional stress results in only marginal deformation [20,21].
Both animal and human trials have confirmed this nonlinear stress–strain relationship between the administered IAP and the gain in intra-abdominal volume (IAV) during pneumoperitoneum. A recent individual patient meta-analysis of patients undergoing laparoscopic surgery analyzed pressure and volume data recorded at the start of insufflation and showed how a non-linear model fitted the pressure–volume (PV) relationship better than a linear model and how a critical point where the abdominal compliance, i.e., the amount of gained volume for an increment in pressure, decreases can be determined by using the function’s first derivative [22]. Similar results were observed by computed tomography (CT) imaging in a porcine animal model of abdominal insufflation, where gains in IAV were shown to be diminishing [23]. Thus, during pneumoperitoneum, the yield stress point is eventually reached where the IAP–IAV curve comes to a plateau; further increases in pressure past such point result in only marginal gains in volume [24]. Of note, the point occurs in the usual range of IAP used for pneumoperitoneum, generally around 10 mmHg, and it can be located by applying non-linear functions such as logistic or exponential to model abdominal behavior during the insufflation [25]. Acknowledging and responding to this fundamental physical principle is crucial, as elevated IAP is associated with various negative effects. In comparison, intra-abdominal hypertension (IAH), i.e., an abdominal pressure above 12 mmHg, is a well-known and acted upon clinical condition in critically ill patients that increases morbidity and mortality through decreased abdominal organ perfusion and increased pressure transmitted to other body organs [26,27,28,29,30,31,32]. Indeed, a raised IAP can displace the diaphragm upward, thus causing a compression of the intrathoracic content that leads to increased transpulmonary pressure and decreased cardiac venous return [33]. This pressure transmission is even more important if body positioning has to be taken into account. For instance, increased intracranial and intraocular pressure has been associated with laparoscopic surgery performed in the Trendelenburg position [34,35,36].
Several studies carried out during pneumoperitoneum insufflation confirm that the pressure increment is associated with morphological and physiological changes to the peritoneal microenvironment that are correlated with the duration of such pressure increment during surgery. For instance, IAP was found to be related to the grade of peritoneal morphological changes such as mesothelial cell bulge and basal lamina exposure in a murine model comparing pneumoperitoneum insufflated at different thresholds compared to control animals submitted to general anesthesia only [37]. In a study carried out on patients undergoing laparoscopic cholecystectomy, the electronic microscopy examination of peritoneal tissue highlighted signs of distress such as edema and cell apoptosis [38]. IAP levels are also associated with the increased release of proinflammatory cytokines and decreased reactive oxygen species scavengers, macrophage activity, and glycocalyx extracellular matrix protein synthesis [39,40]. Moreover, high IAP levels can impair organ perfusion [41]. Both hepatic microcirculation measured by laser Doppler and portal venous flow measured by ultrasound were found to be decreased in patients undergoing laparoscopic cholecystectomy [42,43]. More recent data assessed hepatic blood flow indirectly by the indocyanine green plasma disappearance rate (ICG-PDR) and showed how ICG-PDR was higher and, hence, liver perfusion was lower in patients who underwent colorectal laparoscopy with 12 mmHg of IAP compared to those who received 8 mmHg of IAP [44]. ICG was also employed in another study that assessed peritoneal capillary microcirculation by computerized image analysis in patients who underwent robotic colorectal surgery at 8, 12, or 16 mmHg of IAP. Upon intravenous injection of ICG, the time to reach maximal fluorescent intensity (TMFI) and the maximum fluorescent intensity (MFI) were compared among groups, and shorter TFMI and higher MFI were observed in the low IAP group [45]. Furthermore, cells that are under threat or become damaged release damage-associated molecular patterns (DAMPs); these intracellular molecules normally function as danger signals by binding to toll-like receptors on innate immune cells. In the context of (surgical) trauma and tissue damage, high DAMP levels lead to immune dysregulation resulting in immune suppression and increased susceptibility to infections. [46,47,48].
As for clinical outcomes, several meta-analyses have compared the effect of low IAP on perioperative outcomes in patients undergoing laparoscopic cholecystectomy. A Bayesian network meta-analysis comparing low, i.e., 8 mmHg, standard, i.e., 8–16 mmHg, and high, i.e., over 16 mmHg, IAP showed lower pain scores and shoulder pain at postoperative 24 h in the low IAP arm while no differences were observed in other outcomes such as the need for conversion to open surgery, postoperative nausea and vomiting (PONV) or bleeding, and the duration of surgery or hospital stay [49]. A more recent one with a larger sample size confirmed these results with a high overall quality of evidence according to the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) criteria [50]. Other systematic reviews evaluated the perioperative effect of IAP level during laparoscopy without restricting it to a specific surgical indication. A meta-analysis merging randomized and observational studies compared the effect of low IAP, defined as less than 10 mmHg, with standard IAP, i.e., more than 10 mmHg, during laparoscopy and found lower pain scores in the low IAP ram and no other effect with low quality of evidence [51]. A more recent one that included 85 randomized controlled trials (RCTs) and 7349 subjects compared low (less than 10 mmHg) vs. standard (10 mmHg or higher) IAP during laparoscopy and found that the low IAP arm had a lower incidence of postoperative complications, defined as Clavien–Dindo grade 1 or 2, lower pain scores, lower incidence of PONV, and reduced length of hospital stay without increasing intraoperative adverse events [52]. Accordingly, recent RCTs have linked low IAP during pneumoperitoneum with improved postoperative recovery focusing on the length of hospital stay [53] or postoperative recovery assessed as a patient-reported outcome on a multidimensional score such as the postoperative quality of recovery score (PQRS) [54]. Both RCTs report better outcomes in the low IAP arm.

3. Individualizing Pressure and Volume Measures to Increase the Workspace

Several factors collectively influence the determination of the ideal IAP to balance minimal physiological side effects and an optimal surgical field. These factors include the compliance of the abdominal region, the positioning of the patient during surgery, the depth of neuromuscular blockade, the nature of the surgical procedure, and whether it involves the upper or lower abdomen. Additionally, considerations extend to the procedural complexity, with more intricate surgeries requiring precise adjustments to intra-abdominal pressure for improved visibility and control. The surgeon’s experience is a pivotal aspect, as their skill and expertise contribute to adapting and optimizing IAP based on the specific needs of each surgery. Complicating elements such as intra-abdominal fat, bleeding, or adhesions add an extra layer of complexity. These factors must be carefully evaluated, and IAP should be adjusted accordingly to accommodate these challenges and minimize potential complications.
As previously mentioned, the published evidence shows the beneficial effects of low-pressure pneumoperitoneum at an IAP of <10 mmHg for a wide range of surgical procedures [52]. It should be considered that while IAP levels are important for all the reasons we exposed in the previous section, the actual IAV greatly varies between individuals with the same IAP, and it is the variable that ultimately defines the surgical workspace. Mathematical models show that abdominal compliance can show a mostly linear relationship between pressure and volume up to 12 mmHg but stress the large intra-individual variation in volume at the same level of pressure, illustrated by a difference of a whole liter between two patients in the supine position at an insufflation pressure of 10 mmHg [55]. Indeed, the mean insufflated volume for the same pressure can increase by almost a liter with hip flexion or adopting the Trendelenburg position [55]. A previously published study estimated that the required IAV for an adequate surgical view is around 3L and shows how the IAV at different pressures is greatly influenced by body positioning. The optimized IAV was estimated by analyzing the initial pneumoperitoneum insufflation with low flow of 1–2 L m−1 and drawing a graph between pressure and volume. This study also assessed which factors can be associated with the optimized IAV by fitting a random linear mixed model assessing the effect of age, gender, body mass index (BMI), number of pregnancies, number of previous laparoscopic or open surgeries, type of surgery (right or left hemicolectomy, rectum, or other surgeries), IAV at 15 mmHg of IAP during the insufflation maneuver, and IAP at volume zero. They found that the variables associated with the optimized IAV were IAV at 15 mmHg and IAP at volume zero [56]. An appealing strategy can be to set this volume as the clinical target and use whatever IAP is needed to achieve it as long as the elastic recoil of the abdominal wall is maintained. Although shifting the focus from pressure to the volume of gas required for maintaining an adequate workspace may appear attractive, it is the pressure level that negatively affects peritoneal blood flow, leading to damage, and, consequently, it should be monitored. For instance, capillary perfusion pressure is estimated to drop to 10 mmHg in the distal segments, providing further support for maintaining a threshold of less than 10 mmHg for a minimally invasive pneumoperitoneum during laparoscopy [57]. A sensible course of action could be individualizing treatment and titrating the right IAP level for each surgical case. This approach allows for the dynamic adjustment of pressure based on the specific requirements of the surgeon, patient, and procedure. In fact, although the overall goal is to conduct the surgery with an IAP below 10 mmHg, it is vital to acknowledge that only the surgeon can assess the adequacy of the surgical working field and decide whether an increase in pressure might be necessary. While utilizing low pressure in laparoscopy offers considerable advantages, prioritizing surgical safety remains paramount. It could very well be the case that in patients with poor abdominal compliance in a difficult clinical scenario, such as pelvic resection, more IAP can be needed, but in general, following the European Association for Endoscopic Surgery guidelines should be the guiding principle.
Indeed, several measures have been suggested to increase surgical workspace besides increasing IAP. For instance, pre-stretching of the abdomen has been proposed to achieve an increased IAV. In a porcine model without neuromuscular blockade (NMB) a stepwise peritoneal insufflation maneuver up to 15 mmHg of IAP increased the subsequent IAV and the anteroposterior abdominal diameter reached upon reinsufflation at each 5 mmHg IAP step as measured by computerized tomography (CT) scan [58]. Pre-stretching by raising IAP at 15 mmHg for 5 min when establishing the pneumoperitoneum was implemented as part of a multifaceted strategy for an individualized lower IAP during laparoscopy that yielded clinical benefits compared to standard pressure pneumoperitoneum [54,59]. A metabolic ischemia–reperfusion protective response has also been postulated in addition to this mechanical effect [60,61]. However, the effects of this technique on the peritoneum microenvironment are still unclear. Other studies carried out in humans reported conflicting results on the actual efficacy of this intervention and its marginal effect and interaction with NMB depth [62,63].
Indeed, the optimal depth of NMB for optimal laparoscopic conditions remains an unresolved issue. During moderate NMB, 75% to 90% of receptors in the neuromuscular junction are blocked with an NMB drug, and one to three train-of-four (TOF) responses using neuromuscular monitoring are obtained. Deep NMB is defined by no TOF responses and quantified by at least one post-tetanic count (PTC) response. A PTC of at least one constitutes a deep NMB [64]. There is an abundance of relatively small trials comparing deep with moderate or no NMB, and comparison is complicated as many trials have also applied different IAPs for both intervention arms. A meta-analysis reported a better visual rating as reported by the operating surgeon with deep NMB compared with moderate NMB [65]. Nonetheless, magnetic resonance imaging (MRI) measurements of pneumoperitoneum volumes showed only slight increases from moderate to deep NMB [66]. The additional perceived improvement of the surgical field by the surgeon may be the result of improved surgical stillness and less sudden abdominal contractions that can improve intraoperative safety, especially when operating close to delicate structures [67]. It can be appealing to resort to a deep NMB, especially since the option of reverting it safely is available with sugammadex [68,69,70]. It should be noted that postoperative residual curarization (PORC) is a well-known clinical condition that has been associated with increased postoperative morbidity [71], and it can be observed even after sugammadex reversal [72]. Given the potential for significant differences in how patients respond and the potential drawbacks of a deep NMB, we recommend that low intra-abdominal pressure (IAP) should be supported with at least moderate NMB and the depth of NMB can be increased as needed.
In other words, surgical teams should carefully adjust intra-abdominal pressure (IAP) during laparoscopic procedures, considering factors like the type and complexity of the surgery, their own experience, and the patient’s specific characteristics. However, it’s essential to recognize that raising IAP does not always result in more space or a better surgical view. Surgeons should be mindful not to compromise safety by pursuing lower pressure at the expense of the patient’s well-being. In summary, IAP is a dynamic parameter that surgeons judiciously adjust to optimize the surgical environment while always ensuring the highest level of safety.

4. Temperature and Humidity

Maintaining proper temperature control is essential in surgical procedures as the natural thermoregulation of patients is compromised by the effects of general anesthesia since all hypnotic agents, both intravenous and inhaled, severely disrupt body thermoregulatory function by lowering the point at which defense responses are activated and inhibit vasoconstricting and shivering mechanisms [73]. Intraoperative hypothermia has been linked to several potentially injurious effects. It can alter the hemostatic cascade, since it hampers platelet and clotting factor function [74], thus increasing blood loss and ultimately increasing the need for allogeneic blood transfusion [75]. It can also cause myocardial dysfunction, hypokalemia, and arrythmias [76]. Moreover, it delays both postoperative recovery [77] and wound healing and increases surgical infections and anesthetic drugs’ half-life [78,79,80]. The CO2 for pneumoperitoneum is administered dry and at room temperature and body heat is needed to warm and humidify it to physiologic values. Most dissipated energy is spent to humidify the dry gas rather than warm it. Using standard physical constants, it has been calculated that four times the amount of calories is needed to humidify the same volume of dry gas than warm it from 20 °C to 37 °C [81]. The extent to which this phenomenon might substantially reduce core body temperature remains a topic of debate [82]. Early animal data showed that heating the insufflated CO2 resulted in higher core and peritoneal temperature in a porcine laparoscopic model compared to standard cold insufflated CO2 with a simulated leak of 10 L min−1 [83]. These results were confirmed in a rat model where animals undergoing laparoscopy with heated and humidified CO2 showed higher intraperitoneal and core temperatures than dry or warm, or humidified only insufflated gas [84], resulting in higher core body temperatures [85]. However, these results were not confirmed in a trial carried out in obese patients undergoing laparoscopic gastric bypass where no difference in core temperature was observed between dry and warm and humidified insufflation [86]. Indeed, a recent systematic review only showed slightly higher core temperatures in patients who underwent laparoscopic surgery with warmed and humidified gas compared to the standard dry and cold gas [87], while another showed a slightly higher core temperature with heated and humidified gas compared to a heated-only gas [88]. These conflicting results can probably result in how much and how long the gas is insufflated and the employed alternative warming methods, such as warming blankets and fluid heaters [89]. One recent observational study conducted in a variety of laparoscopic procedures that ranged from appendectomies to colorectal surgery and hernia repair examined the effect of dry and cold insufflation on core body and peritoneal temperature in patients actively warmed by whole body warm air blanket up to 60 min after surgical incision. They observed how core body temperature was unaffected throughout the measurement period while its difference with intraperitoneal temperature increased significantly [90].
Regardless of the systemic consequences, the effect of cold and dry CO2 insufflation on the peritoneal microenvironment should be considered [16]. Animal studies’ data shows that the previously discussed peritoneal injury [39,91,92,93] can be attenuated by humidification. For instance, in an animal RCT comparing the administration of cold and dry versus warm versus humidified insufflated gas, the authors found increased peritoneal injury by electronic microscopy examination and increased intra-abdominal adhesions through direct peritoneal examination two weeks after the laparoscopic intervention [94]. Other authors studying the effect of cold and dry insufflation in mice undergoing laparoscopic surgery observed how abdominal adhesions formation was closely linked to the use of non-humidified gas [95]. There is, however, limited human experimental data regarding peritoneal ultrastructure and the conditions of intraperitoneal temperature and humidity [96]. In an observational study carried out in patients undergoing laparoscopic cholecystectomies, peritoneal biopsies were obtained at the beginning and at the end of the procedure, and the authors report that mesothelial edema, apical cell membrane deterioration, and cell apoptosis can be observed in the latter sample [38]. One contributing factor to this scarcity of data may be the absence of a specialized monitoring system capable of continuously evaluating intra-abdominal temperature and humidity throughout laparoscopy, providing quantitative data since core temperature might not necessarily be an accurate estimation of intraperitoneal temperature. Several commercial devices can humidify and warm the insufflated gas, but, to our knowledge, no intracavitary monitoring system can currently be used as a feedback loop to achieve individualized peritoneal conditioning. Also, the association of the observed peritoneal injury and adhesion formation on clinical outcomes remains to be elucidated.
During laparoscopic procedures, the maintenance of an optimal vision field is key to carrying out the procedure safely and to improve surgical precision and ultimately reduce operative time. One additional benefit of warming and humidifying the insufflated gas is providing a better optical field to the surgeon as laparoscopic lens fogging (LLF) resulting from water condensation can hinder visibility. LLF can also be related to particulate debris, blood, and smoke accumulation on the scope lens [97], and it can increase the surgical time while interfering with the most critical tasks, such as structures’ close-up view, bleeding control, or when there is no haptic feedback, such as in some instances of robotic-assisted surgery. LLF can have various predisposing factors [13,97], although the most accepted theories of its origin revolve around the dew point and psychrometric parameters, i.e., the temperature and humidity of the scope and the surrounding environment. The dew point is the temperature at which condensation happens on a surface, given a specific humidity level and the temperature of the surrounding gas. It has been estimated that to ensure clear vision during laparoscopy, the ambient temperature needs to be above 34 °C when the humidity is around 85%, which is a value that lies to the lower tail of normal laparoscopy. Also, if the temperature of the scope lens exceeds 37.8 °C, there will be no condensation even at 100% humidity. Therefore, it is recommended to keep the scope lens consistently above 37.8 °C to effectively prevent lens fogging [98,99]. Data from animal studies suggest that cooling by convection from the cold insufflation gas caused by the proximity of the injection port to the scope can produce condensation even if it has been pre-warmed [100]. Moreover, if the temperature of the cavity drops below its dew point, fine water droplets in the peritoneal environment can condense into a mist that hinders visibility. This has been related to cold gas insufflation as well [101]. Although these LLF explanations rely on scientific theory and experimental data, extensive comparative analyses on methods of LLF management are still lacking. Indeed, there are even studies that challenge this paradigm [102,103]. All in all, it seems reasonable that administering a warm and humidified gas, especially if accompanied by a supplementary flow, can be beneficial to prevent LLF and, more importantly, preserve peritoneal homeostasis. Indeed, recently developed insufflating systems such as the AirSeal® (ConMed, New York, US) implement an augmented flow to produce gas recirculation to help smoke evacuation and provide a constant clear surgical field that could benefit from additional warmth and humidification [104]. Indeed, in a recent study, the use of this device improved the rate of surgeries that could be carried out at low IAP compared to standard insufflation administration, [105] therefore improving surgeons’ comfort, which can be at times reduced at low IAP [106].

5. Conclusions: Perioperative Goals and Implementation

The introduction of laparoscopic surgery has been one of the greatest advances in perioperative care in the last decades. The enhancement of the insufflation procedure can provide potential benefits to couple with the already implemented advancements in vision and precision. In this review, we revise the existing evidence concerning strategies for maintaining peritoneal homeostasis and averting the systemic and clinical ramifications associated with compromised perfusion and mesothelial cell injury. This can be achieved by (1) an individualized and dynamic adjustment of the intraabdominal pressure, (2) applying measures to increase surgical workspace, and (3) regulating the temperature and humidity of the insufflated gas. Future research should focus on validating this approach further, and insufflators should allow for monitoring and control of IAP, volume, temperature, and humidity.

Author Contributions

Conceptualization, G.M., L.R. and O.D.-C.; methodology, G.M., L.R., K.I.A.-W. and M.C.W.; investigation, G.M. and K.I.A.-W.; writing—original draft preparation, G.M. and L.R.; writing—review and editing, G.M., K.I.A.-W., P.A.-N. and B.F.; visualization, O.D.-C. and P.A.-N.; project administration, G.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The paper does not report any original data.

Conflicts of Interest

G.M., L.R., and O.D.C. are co-inventors on a patent “A medical device for transluminal access” EP20230338055, and have no conflict of interest to declare.

References

  1. The COlon cancer Laparoscopic or Open Resection Study Group. Laparoscopic surgery versus open surgery for colon cancer:short-term outcomes of a randomised trial. Lancet Oncol. 2005, 6, 477–484. [Google Scholar] [CrossRef] [PubMed]
  2. Antoniou, S.A.; Antoniou, G.A.; Koch, O.O.; Pointner, R.; Granderath, F.A. Meta-analysis of laparoscopic vs open cholecystectomy in elderly patients. World J. Gastroenterol. 2014, 20, 17626–17634. [Google Scholar] [CrossRef] [PubMed]
  3. Maas, K.; Biere, S.; Van Der Peet, D.; Cuesta, M. Minimally invasive esophagectomy: Current status and future direction. Surg. Endosc. 2012, 26, 1794. [Google Scholar] [CrossRef] [PubMed]
  4. Spanjersberg, W.R.; van Sambeeck, J.D.P.; Bremers, A.; Rosman, C.; van Laarhoven, C.J.H.M. Systematic review and meta-analysis for laparoscopic versus open colon surgery with or without an ERAS programme. Surg. Endosc. 2015, 29, 3443–3453. [Google Scholar] [CrossRef]
  5. Li, X.; Zhang, J.; Sang, L.; Zhang, W.; Chu, Z.; Liu, Y. Laparoscopic versus conventional appendectomy—A meta-analysis of randomized controlled trials. BMC Gastroenterol. 2010, 10, 129. [Google Scholar] [CrossRef]
  6. Binda, M.M. Humidification during laparoscopic surgery: Overview of the clinical benefits of using humidified gas during laparoscopic surgery. Arch. Gynecol. Obstet. 2015, 292, 955–971. [Google Scholar] [CrossRef]
  7. Reynolds, W. The First Laparoscopic Cholecystectomy. J. Soc. Laparosc. Robot. Surg. 2001, 5, 89–94. [Google Scholar]
  8. Anderson, P.L.; Lathrop, R.A.; Webster, R.J. Robot-like dexterity without computers and motors: A review of hand-held laparoscopic instruments with wrist-like tip articulation. Expert Rev. Med. Devices 2016, 13, 661–672. [Google Scholar] [CrossRef]
  9. Achilli, P.; Mari, G.M.; Crippa, J.; Miranda, A.; Santurro, L.; Riggio, V.; Gerosa, M.; Ascheri, P.; Cordaro, G.; Costanzi, A.T.M.; et al. 4K ultra HD technology reduces operative time and intraoperative blood loss in colorectal laparoscopic surgery. F1000Research 2020, 9, 106. [Google Scholar] [CrossRef]
  10. Dunstan, M.; Smith, R.; Schwab, K.; Scala, A.; Gatenby, P.; Whyte, M.; Rockall, T.; Jourdan, I. Is 3D faster and safer than 4K laparoscopic cholecystectomy? A randomised-controlled trial. Surg. Endosc. 2020, 34, 1729–1735. [Google Scholar] [CrossRef] [PubMed]
  11. Zwart, M.J.W.; Jones, L.R.; Fuente, I.; Balduzzi, A.; Takagi, K.; Novak, S.; Stibbe, L.A.; de Rooij, T.; van Hilst, J.; van Rijssen, L.B.; et al. Performance with robotic surgery versus 3D- and 2D-laparoscopy during pancreatic and biliary anastomoses in a biotissue model: Pooled analysis of two randomized trials. Surg. Endosc. 2022, 36, 4518–4528. [Google Scholar] [CrossRef]
  12. Lanfranco, A.R.; Castellanos, A.E.; Desai, J.P.; Meyers, W.C. Robotic Surgery: A Current Perspective. Ann. Surg. 2004, 239, 14–21. [Google Scholar] [CrossRef]
  13. Schurr, M.O.; Kunert, W.; Arezzo, A.; Buess, G. Review in Minimally Invasive Surgery The Role and Future of Endoscopic Imaging Systems. Endoscopy 1999, 31, 557–562. [Google Scholar] [CrossRef] [PubMed]
  14. Mutsaers, S.E. Mesothelial cells: Their structure, function and role in serosal repair. Respirology 2002, 7, 171–191. [Google Scholar] [CrossRef] [PubMed]
  15. Wilson, R.B. Changes in the coelomic microclimate during carbon dioxide laparoscopy: Morphological and functional implications. Pleura Peritoneum 2017, 2, 17–31. [Google Scholar] [CrossRef] [PubMed]
  16. Umano, G.R.; Delehaye, G.; Noviello, C.; Papparella, A. The “dark Side” of Pneumoperitoneum and Laparoscopy. Minim. Invasive Surg. 2021, 2021. [Google Scholar] [CrossRef] [PubMed]
  17. Papparella, A.; Noviello, C.; Romano, M.; Parmeggiani, P.; Paciello, O.; Papparella, S. Local and systemic impact of pnemoperitoneum on prepuberal rats. Pediatr. Surg. Int. 2007, 23, 453–457. [Google Scholar] [CrossRef] [PubMed]
  18. Neuhaus, S.J.; Gupta, A.; Watson, D.I. Helium and other alternative insufflation gases for laparoscopy: A review. Surg. Endosc. 2001, 15, 553–560. [Google Scholar] [CrossRef]
  19. Neudecker, J.; Sauerland, S.; Neugebauer, E.; Bergamaschi, R.; Bnjer, H.J.; Cuschieri, A.; Fuchs, K.H.; Jacobi, C.; Jansen, F.W.; Koivusalo, A.M.; et al. The European Association for Endoscopic Surgery clinical practice guideline on the pneumoperitoneum for laparoscopic surgery. Surg. Endosc. 2002, 16, 1121–1143. [Google Scholar] [CrossRef] [PubMed]
  20. Becker, C.; Plymale, M.A.; Wennergren, J.; Totten, C.; Stigall, K.; Roth, J.S. Compliance of the abdominal wall during laparoscopic insufflation. Surg. Endosc. 2017, 31, 1947–1951. [Google Scholar] [CrossRef]
  21. Förstemann, T.; Trzewik, J.; Holste, J.; Batke, B.; Konerding, M.A.; Wolloscheck, T.; Hartung, C. Forces and deformations of the abdominal wall-A mechanical and geometrical approach to the linea alba. J. Biomech. 2011, 44, 600–606. [Google Scholar] [CrossRef]
  22. Mazzinari, G.; Diaz-Cambronero, O.; Serpa-Neto, A.; Cañada Martinez, A.; Rovira, L.; Argente-Navarro, M.P.; Malbrain, M.L.N.G.; Pelosi, P.; Gama de Abreu, M.; Hollmann, M.W.; et al. Modeling intra-abdominal volume and respiratory driving pressure during pneumoperitoneum insufflation—A patient-level data meta-analysis. J. Appl. Physiol. 2021, 130, 721–728. [Google Scholar] [CrossRef]
  23. Vlot, J.; Wijnen, R.; Stolker, R.J.; Bax, K. Optimizing working space in porcine laparoscopy: CT measurement of the effects of intra-abdominal pressure. Surg. Endosc. 2013, 27, 1668–1673. [Google Scholar] [CrossRef]
  24. Calvo, G.; Armero, C.; Gómez-Rubio, V.; Mazzinari, G. Bayesian hierarchical nonlinear modelling of intra-abdominal volume during pneumoperitoneum for laparoscopic surgery. SORT 2021, 45, 1–20. [Google Scholar] [CrossRef]
  25. Archontoulis, S.V.; Miguez, F.E. Nonlinear regression models and applications in agricultural research. Agron. J. 2015, 107, 786–798. [Google Scholar] [CrossRef]
  26. De Laet, I.E.; Malbrain, M.L.N.G.; De Waele, J.J. A Clinician’s Guide to Management of Intra-Abdominal Hypertension and Abdominal Compartment Syndrome in Critically Ill Patients. Crit. Care 2020, 24, 1–9. [Google Scholar] [CrossRef] [PubMed]
  27. Malbrain, M.L.N.G.; Roberts, D.J.; Sugrue, M.; De Keulenauer, B.L.; Ivatury, R.; Pelosi, P.; Verbrugge, F.; Wise, R.; Mullens, W. The polycompartment syndrome: A concise state-of-the-art review. Anaesthesiol. Intensive Ther. 2014, 46, 433–450. [Google Scholar] [CrossRef]
  28. Van Damme, L.; De Waele, J.J. Effect of decompressive laparotomy on organ function in patients with abdominal compartment syndrome: A systematic review and meta-analysis. Crit. Care 2018, 22, 179. [Google Scholar] [CrossRef] [PubMed]
  29. Wever, K.E.; Bruintjes, M.H.D.; Warlé, M.C.; Hooijmans, C.R. Renal perfusion and function during pneumoperitoneum: A systematic review and meta-analysis of animal studies. PLoS ONE 2016, 11, e0163419. [Google Scholar] [CrossRef] [PubMed]
  30. Zhou, J.C.; Xu, Q.P.; Pan, K.H.; Mao, C.; Jin, C.W. Effect of increased intra-abdominal pressure and decompressive laparotomy on aerated lung volume. J. Zhejiang Univ. Sci. B 2010, 11, 378–385. [Google Scholar] [CrossRef] [PubMed]
  31. Mazzinari, G.; Diaz-Cambronero, O.; Alonso-Iñigo, J.M.; García-Gregorio, N.; Ayas_Montero, B.; Ibañez, J.L.; Serpa-Neto, A.; Ball, L.; Gama de Abreu, M.; Pelosi, P.; et al. Intraabdominal pressure targeted positive end-expiratory pressure during laparoscopic surgery: An open-label, nonrandomized, crossover, clinical trial. Anesthesiology 2020, 132, 667–677. [Google Scholar] [CrossRef] [PubMed]
  32. Cheng, M.; Ni, L.; Huang, L.; Zhou, Y.; Wang, K. Effect of positive end-expiratory pressure on pulmonary compliance and pulmonary complications in patients undergoing robot-assisted laparoscopic radical prostatectomy: A randomized control trial. BMC Anesthesiol. 2022, 22, 347. [Google Scholar] [CrossRef] [PubMed]
  33. Valenza, F.; Chevallard, G.; Fossali, T.; Salice, V.; Pizzocri, M.; Gattinoni, L. Management of mechanical ventilation during laparoscopic surgery. Best Pract. Res. Clin. Anaesthesiol. 2010, 24, 227–241. [Google Scholar] [CrossRef] [PubMed]
  34. Awad, H.; Santilli, S.; Ohr, M.; Roth, A.; Yan, W.; Fernandez, S.; Roth, S.; Patel, V. The effects of steep trendelenburg positioning on intraocular pressure during robotic radical prostatectomy. Anesth. Analg. 2009, 109, 473–478. [Google Scholar] [CrossRef] [PubMed]
  35. Robba, C.; Cardim, D.; Donnelly, J.; Bertuccio, A.; Bacigaluppi, S.; Bragazzi, N.; Cabella, B.; Liu, X.; Matta, B.; Lattuada, M.; et al. Effects of pneumoperitoneum and Trendelenburgposition on intracranial pressure assessed using different non-invasive methods. Br. J. Anaesth. 2016, 117, 783–791. [Google Scholar] [CrossRef] [PubMed]
  36. Kalmar, A.F.; Foubert, L.; Hendrickx, J.F.A.; Mottrie, M.; Absalom, A.; Mortier, E.P.; Struys, M.M.R.F. Influence of steep Trendelenburg position and CO2 pneumoperitoneum on cardiovascular, cerebrovascular, and respiratory homeostasis during robotic prostatectomy. Br. J. Anaesth. 2010, 104, 433–439. [Google Scholar] [CrossRef] [PubMed]
  37. Papparella, A.; Nino, F.; Coppola, S.; Noviello, C.; Paciello, O.; Papparella, S. Peritoneal morphological changes due to pneumoperitoneum: The effect of intra-abdominal pressure. Eur. J. Pediatr. Surg. 2014, 24, 322–327. [Google Scholar] [CrossRef] [PubMed]
  38. Tarhan, O.R.; Barut, I.; Ozogul, C.; Bozkurt, S.; Baykara, B.; Bulbul, M. Structural deteriorations of the human peritoneum during laparoscopic cholecystectomy. A transmission electron microscopic study. Surg. Endosc. 2013, 27, 2744–2750. [Google Scholar] [CrossRef]
  39. Neuhaus, S.J.; Watson, D.I. Pneumoperitoneum and peritoneal surface changes—A review. Surg. Endosc. 2004, 18, 1316–1322. [Google Scholar] [CrossRef]
  40. Matsuzaki, S.; Jardon, K.; Maleysson, E.; Darpiany, F.; Canis, M.; Botchorishvili, R. Impact of intraperitoneal pressure of a CO2 pneumoperitoneum on the surgical peritoneal environment. Hum. Reprod. 2012, 27, 1613–1623. [Google Scholar] [CrossRef]
  41. Sammour, T.; Mittal, A.; Loveday, B.P.T.; Kahokehr, A.; Phillips, A.R.J.; Windsor, J.A.; Hill, A.G. Systematic review of oxidative stress associated with pneumoperitoneum. Br. J. Surg. 2009, 96, 836–850. [Google Scholar] [CrossRef]
  42. Eleftheriadis, E.; Kotzampassi, K.; Botsios, D.; Tzartinoglou, E.; Farmakis, H.; Dadoukis, J. Splanchnic Ischemia during Laparoscopic Cholecystectomy. Surg. Endosc. 1996, 10, 324–326. [Google Scholar] [CrossRef]
  43. Jakimowicz, J.; Stultiëns, G.; Smulders, F. Laparoscopic Insufflation of the Abdomen Reduces Portal Venous Flow. Surg. Endosc. 1998, 12, 129–132. [Google Scholar] [CrossRef] [PubMed]
  44. Olmedilla Arnal, L.E.; Cambronero, O.D.; Mazzinari, G.; Perez Peña, J.M.; Zorrilla Ortuzar, J.; Rodriguez Martin, M.; Vila Montañes, M.; Schultz, M.J.; Rovira, L.; Navarro , M.P.A.; et al. An Individualized Low-Pneumoperitoneum-Pressure Strategy May Prevent a Reduction in Liver Perfusion during Colorectal Laparoscopic Surgery. Biomedicines 2023, 11, 891. [Google Scholar] [CrossRef] [PubMed]
  45. Albers, K.I.; Polat, F.; Loonen, T.; Graat, L.J.; Mulier, J.P.; Snoeck, M.M.J.; Pnahuizen, I.F.; Vermulst, A.A.; Scheffer, G.J.; Warlé, M.C. Visualising improved peritoneal perfusion at lower intra-abdominal pressure by fluorescent imaging during laparoscopic surgery: A randomised controlled study. Int. J. Surg. 2020, 77, 8–13. [Google Scholar] [CrossRef] [PubMed]
  46. Sharma, S.K.; Naidu, G. The role of danger-associated molecular patterns (DAMPs) in trauma and infections. J. Thorac. Dis. 2016, 8, 1406–1409. [Google Scholar] [CrossRef] [PubMed]
  47. Relja, B.; Land, W.G. Damage-associated molecular patterns in trauma. Eur. J. Trauma Emerg. Surg. 2020, 46, 751–775. [Google Scholar] [CrossRef] [PubMed]
  48. Leijte, G.P.; Custers, H.; Gerretsen, J.; Heijne, A.; Roth, J.; Vogl, T.; Scheffer, G.J.; Pickkers, P.; Kox, M. Increased plasma levels of danger-associated molecular patterns are associated with immune suppression and postoperative infections in patients undergoing cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Front. Immunol. 2018, 9, 663. [Google Scholar] [CrossRef]
  49. Raval, A.D.; Deshpande, S.; Koufopoulou, M.; Rabar, S.; Neupane, B.; Iheanacho, I.; Bash, L.D.; Horrow, J.; Fuchs-Buder, T. The impact of intra-abdominal pressure on perioperative outcomes in laparoscopic cholecystectomy: A systematic review and network meta-analysis of randomized controlled trials. Surg. Endosc. 2020, 34, 2878–2890. [Google Scholar] [CrossRef]
  50. Ortenzi, M.; Montori, G.; Sartori, A.; Balla, A.; Botteri, E.; Piatto, G.; Gallo, G.; Vigna, S.; Guerrieri, M.; Williams, S.; et al. Low-pressure versus standard-pressure pneumoperitoneum in laparoscopic cholecystectomy: A systematic review and meta-analysis of randomized controlled trials. Surg. Endosc. 2022, 36, 7092–7113. [Google Scholar] [CrossRef]
  51. Özdemir-van Brunschot, D.M.D.; van Laarhoven, K.C.J.H.M.; Scheffer, G.J.; Pouwels, S.; Wever, K.E.; Warlé, M.C. What is the evidence for the use of low-pressure pneumoperitoneum? A systematic review. Surg. Endosc. 2016, 30, 2049–2065. [Google Scholar] [CrossRef] [PubMed]
  52. Reijnders-Boerboom, G.T.J.A.; Albers, K.I.; Jacobs, L.M.C.; van Helden, E.; Rsoman, C.; Díaz-Cambronero, O.; Mazzinari, G.; Scheffer, G.J.; Keijzer, C.; Warleé, M.C. Low intra-abdominal pressure in laparoscopic surgery: A systematic review and meta-analysis. Int. J. Surg. 2023, 109, 1400–1411. [Google Scholar] [CrossRef] [PubMed]
  53. Celarier, S.; Monziols, S.; Célérier, B.; Assenat, V.; Carles, P.; Napolitano, G.; Laclau-Lacrouts, M.; Rullier, E.; Ouattara, A.; Denost, Q. Low-pressure versus standard pressure laparoscopic colorectal surgery (PAROS trial): A phase III randomized controlled trial. Br. J. Surg. 2021, 108, 998–1005. [Google Scholar] [CrossRef] [PubMed]
  54. Díaz-Cambronero, O.; Mazzinari, G.; Flor Lorente, B.; García Gregorio, N.; Robles-Hernandez, D.; Olmedilla Arnal, L.; Martin de Pablos, A.; Schultz, M.J.; Errando, C.L.; Argente Navarro, M.P.; et al. Effect of an individualized versus standard pneumoperitoneum pressure strategy on postoperative recovery: A randomized clinical trial in laparoscopic colorectal surgery. Br. J. Surg. 2020, 107, 1605–1614. [Google Scholar] [CrossRef]
  55. Mulier, J.; Dillemans, B.; Crombach, M.; Missant, C.; Sels, A. On the abdominal pressure volume relationship. Internet J. Anesthesiol. 2008, 21, 5221–5231. [Google Scholar]
  56. Diaz-Cambronero, O.; Flor Lorente, B.; Mazzinari, G.; Vila Montañes, M.; García Gregorio, N.; Robles Hernandez, D.; Olmedilla Arnal, L.E.; Argetne Navarro, M.P.; Schultz, M.J.; Errando, C.L.; et al. A multifaceted individualized pneumoperitoneum strategy for laparoscopic colorectal surgery: A multicenter observational feasibility study. Surg. Endosc. 2019, 33, 252–260. [Google Scholar] [CrossRef] [PubMed]
  57. Solass, W.; Horvath, P.; Struller, F.; Konigsrainer, I.; Beckert, S.; Konigsrainer, A.; Weinreich, F.J.; Schenk, M. Functional vascular anatomy of the peritoneum in health and disease. Pleura Peritoneum 2016, 1, 145–158. [Google Scholar] [CrossRef]
  58. Vlot, J.; Wijnen, R.; Stolker, R.J.; Bax, K. Optimizing working space in laparoscopy: CT measurement of the effect of pre-stretching of the abdominal wall in a porcine model. Surg. Endosc. 2014, 28, 841–846. [Google Scholar] [CrossRef]
  59. Diaz-Cambronero, O.; Mazzinari, G.; Errando, C.L.; Schult, M.J.; Flor Lorente, B.; García-Gregorio, N.; Vila Montañes, M.; Robles; Olmedilla Arnal, L.E.; Martin de-Pablos, A.; et al. An individualised versus a conventional pneumoperitoneum pressure strategy during colorectal laparoscopic surgery: Rationale and study protocol for a multicentre randomised clinical study. Trials 2019, 20, 190. [Google Scholar] [CrossRef]
  60. Biler, A.; Yucebilgin, S.; Sendag, F.; Akman, L.; Akdemir, A.; Ates, U.; Uyianigkil, Y.; Yilmaz-Dilsiz, O.; Sezer, E. The Effects of Different Intraabdominal Pressure Protocols in Laparoscopic Procedures on Oxidative Stress Markers and Morphology in Rat Ovaries. Adv. Clin. Exp. Med. 2014, 2014, 885–892. [Google Scholar] [CrossRef]
  61. Cevrioglu, A.S.; Yilmaz, S.; Koken, T.; Tokyol, C.; Yilmazer, M.; Fenkci, I.V. Comparison of the effects of low intra-abdominal pressure and ischaemic preconditioning on the generation of oxidative stress markers and inflammatory cytokines during laparoscopy in rats. Hum. Reprod. 2004, 19, 2144–2151. [Google Scholar] [CrossRef]
  62. Mulier, J.P.J.; Dillemans, B.; Van Cauwenberge, S. Impact of the patient’s body position on the intraabdominal workspace during laparoscopic surgery. Surg. Endosc. 2010, 24, 1398–1402. [Google Scholar] [CrossRef]
  63. Barrio, J.; Errando, C.L.; San Miguel, G.; Salas, B.I.; Raga, J.; Carrión, J.L.; García-Ramón, J.; Gallego, J. Effect of depth of neuromuscular blockade on the abdominal space during pneumoperitoneum establishment in laparoscopic surgery. J. Clin. Anesth. 2016, 34, 197–203. [Google Scholar] [CrossRef]
  64. Fuchs-Buder, T.; Schreiber, J.U.; Meistelman, C. Monitoring neuromuscular block: An update. Anaesthesia 2009, 64 (Suppl. S1), 82–89. [Google Scholar] [CrossRef]
  65. Martini, C.H.; Boon, M.; Bevers, R.F.; Aarts, L.P.; Dahan, A. Evaluation of surgical conditions during laparoscopic surgery in patients with moderate vs deep neuromuscular block. Br. J. Anaesth. 2014, 112, 498–505. [Google Scholar] [CrossRef]
  66. Krijtenburg, P.; Bruintjes, M.H.D.; Fütterer, J.J.; van de Steeg, G.; d’Ancona, F.; Scheffer, G.J.; Keijzer, C.; Warlé, M.C. MRI measurement of the effects of moderate and deep neuromuscular blockade on the abdominal working space during laparoscopic surgery, a clinical study. BMC Anesthesiol. 2023, 23, 238. [Google Scholar] [CrossRef] [PubMed]
  67. Rosenberg, J.; Fuchs-Buder, T. Surgical Stillness—When, Why, and How? Front. Surg. 2019, 6, 61. [Google Scholar] [CrossRef] [PubMed]
  68. Hristovska, A.M.; Duch, P.; Allingstrup, M.; Afshari, A. Efficacy and safety of sugammadex versus neostigmine in reversing neuromuscular blockade in adults. Cochrane Database Syst. Rev. 2017, 8, CD012763. [Google Scholar] [CrossRef] [PubMed]
  69. Raval, A.D.; Anupindi, V.R.; Ferrufino, C.P.; Arper, D.L.; Bash, L.D.; Brull, S.J. Epidemiology and outcomes of residual neuromuscular blockade: A systematic review of observational studies. J. Clin. Anesth. 2020, 66, 109962. [Google Scholar] [CrossRef]
  70. Kotake, Y.; Ochiai, R.; Suzuki, T.; Ogawa, S.; Takagi, S.; Ozaki, M.; Nakatsuka, I.; Takeda, J. Reversal with sugammadex in the absence of monitoring did not preclude residual neuromuscular block. Anesth. Analg. 2013, 117, 345–351. [Google Scholar] [CrossRef] [PubMed]
  71. Carvalho, H.; Verdonck, M.; Cools, W.; Geerts, L.; Forget, P.; Poelaert, J. Forty years of neuromuscular monitoring and postoperative residual curarisation: A meta-analysis and evaluation of confidence in network meta-analysis. Br. J. Anaesth. 2020, 125, 466–482. [Google Scholar] [CrossRef] [PubMed]
  72. Díaz-Cambronero, Ó.; Mazzinari, G.; Errando, C.L.; Garutti, I.; Abad-Gurumeta, A.; Serrano, A.B.; Esteve, N.; Montañes, M.V.; Neto, A.S.; Hollmann, M.W.; et al. An educational intervention to reduce the incidence of postoperative residual curarisation: A cluster randomised crossover trial in patients undergoing general anaesthesia. Br. J. Anaesth. 2023, 131, 482–490. [Google Scholar] [CrossRef] [PubMed]
  73. Sessler, D.I. Perioperative Temperature Monitoring. Anesthesiology 2021, 134, 111–118. [Google Scholar] [CrossRef] [PubMed]
  74. Hardy, J.F.; de Moerloose, P.; Samama, C.M.; Members of the Groupe d’Intérêt en Hémostase Périopératoire. Massive transfusion and coagulopathy: Pathophysiology and implications for clinical management. Can. J. Anesth. 2006, 53, S40–S58. [Google Scholar] [CrossRef]
  75. Schmied, H.; Kurz, A.; Sessler, D.I.; Kozek, S.; Reiter, A. Mild hypothermia increases blood loss and transfusion requirements during total hip arthroplasty. Lancet 1996, 347, 289–292. [Google Scholar] [CrossRef] [PubMed]
  76. Frank, S.M.; Fleisher, L.A.; Breslow, M.J.; Higins, M.S.; Olson, K.F.; Beattie, C. Perioperative Maintenance of Normothermia Reduces the Incidence of Morbid Cardiac Events A Randomized Clinical Trial. JAMA 1997, 277, 1127–1134. [Google Scholar] [CrossRef] [PubMed]
  77. Sessler, D.I.; Sessler, A.M. Experimental determination of heat flow parameters during induction of general anesthesia. Anesthesiology 1998, 89, 657–665. [Google Scholar] [CrossRef] [PubMed]
  78. Kurz, A.; Sessler, D.I.; Lenhardt, R. Perioeperative normothermia to reduce the incidence of surgical-wound infection and shorten hospitalization. NEJM 1996, 334, 1209–1215. [Google Scholar] [CrossRef]
  79. Melling, A.C.; Ali, B.; Scott, E.M.; Leaper, D.J. Effects of preoperative warming on the incidence of wound infection after clean surgery: A randomised controlled trial. Lancet 2001, 358, 876–880. [Google Scholar] [CrossRef]
  80. Leslie, K.; Sessler, D.I.; Bjorksten, A.R.; Moayeri, A. Mild Hypothermia Alters Propofol Pharmacokinetics and Increases the Duration of Action of Atracurium. Anesth. Analg. 1995, 80, 1007–1021. [Google Scholar] [CrossRef]
  81. Roth, J.V.; Sea, S.; Laparosc, S.; Percutan, E. An Assessment by Calorimetric Calculations of the Potential Thermal Benefit of Warming and Humidification of Insufflated Carbon Dioxide. Surg. Laparosc. Endosc. Percutaneous Tech. 2014, 24, e106. [Google Scholar] [CrossRef]
  82. De Csepel, J.; Wilson, E. Heating and humidifying carbon dioxide is indicated. Surg. Endosc. 2007, 21, 340–341. [Google Scholar] [CrossRef]
  83. Besseil, J.R.; Karatassas, A.; Patterson, J.R.; Jamieson, G.G.; Maddern, G.J. Hypothermia Induced by Laparoscopic Insufflation A Randomized Study in a Pig Model. Surg. Endosc. 1995, 9, 791–796. [Google Scholar] [CrossRef]
  84. Hazerbroek, E.; Schreve, M.; Visser, P.; De Bruin, R.; Marquet, R.; Bonjer, H. Impact of Temperature and Humidity of Carbon Dioxide Pneumoperitoneum on Body Temperature and Peritoneal Morphology. J. Laparoendosc. Adv. Surg. Tech. 2002, 12, 355–364. [Google Scholar] [CrossRef]
  85. Mouton, W.G.; Bessell, J.R.; Pfitzner, J.; Dymock, R.B.; Brealey, J.; Maddern, G.J. A Randomized Controlled Trial to Determine the Effects of Humidified Carbon Dioxide Insufflation during Thoracoscopy. Surg. Endosc. 1999, 13, 382–385. [Google Scholar] [CrossRef] [PubMed]
  86. Davis, S.S.; Mikami, D.J.; Newlin, M.; Needleman, B.J.; Barret, M.S.; Fries, R.; Larson, T.; Dundon, J.; Goldblatt, M.I.; Melvin, W.S. Heating and humidifying of carbon dioxide during pneumoperitoneum is not indicated: A prospective randomized trial. Surg. Endosc. 2006, 20, 153–158. [Google Scholar] [CrossRef] [PubMed]
  87. Balayssac, D.; Pereira, B.; Bazin, J.E.; Roy BLe Pezet, D.; Gagnière, J. Warmed and humidified carbon dioxide for abdominal laparoscopic surgery: Meta-analysis of the current literature. Surg. Endosc. 2017, 31, 1–12. [Google Scholar] [CrossRef] [PubMed]
  88. Birch, D.W.; Dang, J.T.; Switzer, N.J.; Manoucheheri, N.; Shi, X.; Hadi, G.; Karmali, S. Heated insufflation with or without humidification for laparoscopic abdominal surgery. Cochrane Database Syst. Rev. 2016, 2016, CD007821. [Google Scholar] [CrossRef] [PubMed]
  89. Dean, M.; Ramsay, R.; Heriot, A.; Mackay, J.; Hiscock, R.; Lynch, A.C. Warmed, humidified CO2 insufflation benefits intraoperative core temperature during laparoscopic surgery: A meta-analysis. Asian J. Endosc. Surg. 2017, 10, 128–136. [Google Scholar] [CrossRef] [PubMed]
  90. Groene, P.; Gündogar, U.; Hofmann-Kiefer, K.; Ladurner, R. Influence of insufflated carbon dioxide on abdominal temperature compared to oesophageal temperature during laparoscopic surgery. Surg. Endosc. 2021, 35, 6892–6896. [Google Scholar] [CrossRef] [PubMed]
  91. Wong, Y.T.; Shah, P.C.; Birkett, D.H.; Brams, D.M. Peritoneal pH during laparoscopy is dependent on ambient gas environment: Helium and nitrous oxide do not cause peritoneal acidosis. Surg. Endosc. 2005, 19, 60–64. [Google Scholar] [CrossRef]
  92. Taskin, O.; Buhur, A.; Birincioglu, M.; Burak, F.; Atmaca, R.; Yilmaz, I.; Wheeler, J.M. The Effects of Duration of CO2 Insufflation and Irrigation on Peritoneal Microcirculation Assessed by Free Radical Scavengers andTotal Glutathion Levels During Operative Laparoscopy. J. Am. Assoc. Gynecol. Laparosc. 1998, 5, 129–133. [Google Scholar] [CrossRef] [PubMed]
  93. Yesildaglar, N.; Koninckx, P.R.; Gasthuisberg, H. Adhesion formation in intubated rabbits increases with high insufflation pressure during endoscopic surgery Adhesions are a major cause of infertility. Hum. Reprod. 2000, 15, 687–691. [Google Scholar] [CrossRef] [PubMed]
  94. Peng, Y.; Zheng, M.; Ye, Q.; Chen, X.; Yu, B.; Liu, B. Heated and Humidified CO2 Prevents Hypothermia, Peritoneal Injury, and Intra-Abdominal Adhesions During Prolonged Laparoscopic Insufflations. J. Surg. Res. 2009, 151, 40–47. [Google Scholar] [CrossRef]
  95. Binda, M.M.; Molinas, C.R.; Hansen, P.; Koninckx, P.R. Effect of desiccation and temperature during laparoscopy on adhesion formation in mice. Fertil. Steril. 2006, 86, 166–175. [Google Scholar] [CrossRef]
  96. Ott, D.E. Correction of Laparoscopic Insufflation Hypothermia. J. Laparoendosc. Surg. 1991, 1, 183–186. [Google Scholar] [CrossRef] [PubMed]
  97. Lawrentschuk, N.; Fleshner, N.E.; Bolton, D.M. Laparoscopic Lens Fogging: A Review of Etiology and Methods to Maintain a Clear Visual Field. J. Endourol. 2010, 24, 905–913. [Google Scholar] [CrossRef]
  98. Bessell, J.R.; Flemming, E.; Kunert, W.; Buess, G. Maintenance of clear vision during laparoscopic surgery. Minim. Invasive Ther. Allied Technol. 1996, 5, 450–455. [Google Scholar] [CrossRef]
  99. Flemming, E.; Bessell, J.R.; Kunert, W.; Eibl, H.; Buess, G. Principles determining optical clarity in endoscopic surgery. Minim. Invasive Ther. Allied Technol. 1996, 5, 440–444. [Google Scholar] [CrossRef]
  100. Tittel, A.; Schippers, E.; Grablowitz, V.; Pollivoda, M.; Anurov, M.; Ottinger, A.; Schumpelick, V. Intraabdominal humidity and electromyographic activity of the gastrointestinal tract Laparoscopy versus laparotomy. Surg. Endosc. 1995, 9, 786–790. [Google Scholar] [CrossRef]
  101. Manning, T.G.; Perera, M.; Christidis, D.; Kinnear, N.; McGrath, S.; O’Beirne, R.; Zotov, P.; Bolton, D.; Lawrentschuck, N. Visual occlusion during minimally invasive surgery: A contemporary review of methods to reduce laparoscopic and robotic lens fogging and other sources of optical loss. J. Endourol. 2017, 31, 327–333. [Google Scholar] [CrossRef]
  102. Nguyen, N.T.; Furdui, G.; Fleming, N.W.; Lee, S.J.; Goldman, C.D.; Singh, A.; Wolfe, B.M. Effect of heated and humidified carbon dioxide gas on core temperature and postoperative pain: A randomized trial. Surg. Endosc. 2002, 16, 1050–1054. [Google Scholar] [CrossRef]
  103. Farley, D.R.; Greenlee, S.M.; Larson, D.R.; Harrington, J.R. Double-blind, Prospective, Randomized Study of Warmed, Humidified Carbon Dioxide Insufflation vs Standard Carbon Dioxide for Patients Undergoing Laparoscopic Cholecystectomy. Arch. Surg. 2004, 139, 739–744. [Google Scholar] [CrossRef] [PubMed]
  104. Ayoub, C.H.; Armache, A.K.; El-Asmar, J.M.; El-Achkar, A.; Abdulfattah, S.; Bidikian, S.; Chawareb, E.A.; Hoyek, E.; El-Hajj, A. The impact of AirSeal® on complications and pain management during robotic-assisted radical prostatectomy: A single-tertiary center study. World J. Urol. 2023, 41, 2685–2692. [Google Scholar] [CrossRef] [PubMed]
  105. Saway, J.P.; McCaul, M.; Mulekar, M.S.; McMahon, D.P.; Richards, W.O. Review of outcomes of low verses standard pressure pneumoperitoneum in laparoscopic surgery. Am. Surg. 2022, 88, 1832–1837. [Google Scholar] [CrossRef] [PubMed]
  106. Vijayaraghavan, N.; Sistla, S.C.; Kundra, P.; Ananthanarayan, P.H.; Karthikeyan, V.S.; Ali, S.M.; Sasi, S.P.; Vikram, K. Comparison of Standard-pressure and Low-pressure Pneumoperitoneum in Laparoscopic Cholecystectomy: A Double Blinded Randomized Controlled Study. Surg. Laparosc. Endosc. Percutaneous Tech. 2014, 24, 127–133. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mazzinari, G.; Rovira, L.; Albers-Warlé, K.I.; Warlé, M.C.; Argente-Navarro, P.; Flor, B.; Diaz-Cambronero, O. Underneath Images and Robots, Looking Deeper into the Pneumoperitoneum: A Narrative Review. J. Clin. Med. 2024, 13, 1080. https://doi.org/10.3390/jcm13041080

AMA Style

Mazzinari G, Rovira L, Albers-Warlé KI, Warlé MC, Argente-Navarro P, Flor B, Diaz-Cambronero O. Underneath Images and Robots, Looking Deeper into the Pneumoperitoneum: A Narrative Review. Journal of Clinical Medicine. 2024; 13(4):1080. https://doi.org/10.3390/jcm13041080

Chicago/Turabian Style

Mazzinari, Guido, Lucas Rovira, Kim I. Albers-Warlé, Michiel C. Warlé, Pilar Argente-Navarro, Blas Flor, and Oscar Diaz-Cambronero. 2024. "Underneath Images and Robots, Looking Deeper into the Pneumoperitoneum: A Narrative Review" Journal of Clinical Medicine 13, no. 4: 1080. https://doi.org/10.3390/jcm13041080

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop