Complement therapeutic strategies in trauma, hemorrhagic shock and systemic inflammation – closing Pandora’s box?

https://doi.org/10.1016/j.smim.2016.04.005Get rights and content

Highlights

  • Severe tissue damage and activation of the coagulation system may trigger the complement cascade through established and non-canonical pathways.

  • Severe tissue trauma and hemorrhagic shock leads to complement activation and complementopathy.

  • Upstream inhibition of the complement cascade at the level of C3 may reduce trauma- and hemorrhagic shock-induced organ damage and improve overall outcome.

  • Downstream specific blockade of the C5a-C5aR interaction may especially ameliorate septic conditions and complications following severe trauma and hemorrhagic shock.

Abstract

After severe trauma, the immune system is challenged with a multitude of endogenous and exogenous danger molecules. The recognition of released danger patterns is one of the prime tasks of the innate immune system. In the last two decades, numerous studies have established the complement cascade as a major effector system that detects and processes such danger signals. Animal models with engineered deficiencies in certain complement proteins have demonstrated that widespread complement activation after severe injury culminates in complement dysregulation and excessive generation of complement activation fragments. Such exuberant pro-inflammatory signaling evokes systemic inflammation, causes increased susceptibility to infections and is associated with a detrimental course of the disease after injury. We discuss the underlying processes of such complementopathy and recapitulate different intervention strategies within the complement cascade. So far, several orthogonal anti-complement approaches have been tested with varying success in a large number of rodent, in several porcine and few simian studies. We illustrate the different features among those intervention strategies and highlight those that hold the greatest promise to become potential therapeutic options for the intricate disease of traumatic injury.

Introduction

In analogy to Pandora’s box that floods the world with evil, severe tissue trauma and hemorrhagic shock overflow the injured body with vast amounts of damaged tissue and inflammatory mediators. So far, no means seem to exist to stop this fatal process. Trauma-hemorrhage exposes the body to various danger- and pathogen-associated molecular patterns (DAMPs and PAMPs, resp.) [1], often resulting in a systemic inflammatory reaction through an immediate release of pro- and anti-inflammatory mediators [2], [3]. Clinically, this generalized inflammation is known as “systemic inflammatory response syndrome” (SIRS). SIRS is diagnosed when at least two of the following criteria are met: (i) temperature of more than 38 °C or less than 36 °C; (ii) heart rate of more than 90 beats per minute; (iii) respiratory rate of more than 20 breaths per minute or arterial carbon dioxide tension (PaCO2) of less than 32 mmHg; (iv) abnormal white blood cell count (>12,000/μL or <4000/μL or >10% immature [band] forms) [4]. Whenever SIRS is associated with a bacterial infection, this inflammatory syndrome is classified as sepsis. Pathomechanistic insights of how such a generalized inflammation is initiated and propagated in response to trauma or traumatic-hemorrhagic shock still remain to be elucidated in detail.

Soon after trauma, exposure to PAMPs/DAMPs extensively challenges the “first line of defense” which comprises leukocytes [3] and the two cross-talking serine protease systems of the coagulation and complement cascade [5], [6]. Especially activation of the complement system quickly senses damaged tissue and danger molecules with attachment of C1qrs and C3b opsonisation, and generation of the anaphylatoxins C3a and C5a translates these signals into an effective cellular danger response [7]. The subsequent cellular oxidative burst reaction leads to release of reactive oxygen radicals (ROS) and tissue- and barrier-degrading proteases from neutrophils (PMN) which, together with vasoactive mediators (e.g. ROS, thrombin), increase the permeability of membranes and the microvasculature. Complement activation products also trigger extensive release of pro-inflammatory cytokines that in turn contribute to systemic inflammation and generalized capillary leakage. The resultant fluid shift into the interstitial space leads to tissue edema and development of multi-organ dysfunction syndrome (MODS) and is responsible for the high mortality rates that occur in later stages after severe trauma (Fig. 1). Organ or multiple organ failure with incidences of 37.2% and 22.1%, respectively, are indeed the most severe complications after trauma and are associated with a mortality rate of at least 70%, thus even outranging the mortality observed for septic complications after trauma (TraumaRegister DGU® 2012).

Another crucial risk factor for a poor outcome after severe tissue trauma is the occurrence of hemorrhagic shock [8] which is defined as a life-threatening reduction in blood perfusion of tissues due to a significant blood loss and ultimately results in further cellular and organ injury. Treatment of coagulopathy after severe hemorrhagic or trauma/hemorrhagic shock with massive blood transfusion is associated with a higher risk of multi-organ failure and death [9].

Despite improved rescue protocols, minimal invasive surgical and modern intensive care treatment, effective and reliable strategies to monitor and control the systemic inflammatory response and associated organ dysfunctions after severe trauma and hemorrhagic shock are still missing in the clinic despite of being desperately needed.

Section snippets

Complement activation and complementopathy after trauma, hemorrhagic shock and during systemic inflammation

Soon after polytraumatic injury in conjunction with hemorrhagic shock, extensive complement activation has been demonstrated in humans, culminating in depletion of crucial complement proteins, which can be monitored clinically by determining the serum complement hemolytic activity (CH50) [10]. Remarkably, also in a rodent model of pressure-controlled hemorrhagic shock (mean arterial pressure 30–35 mmHg for 120 min), CH50 values significantly drop soon after shock induction. Of note, CH50 values

Complement inhibition after trauma: risks and chances

During the last years, numerous studies have identified complement as the predominant mediator of local and systemic inflammatory processes that ultimately lead to organ damage and failure [35]. Using modulatory or inhibitory approaches, many studies have attempted to interfere therapeutically in rodent models of isolated traumatic brain injury (TBI) by inhibiting complement activation at almost all levels possible (C1r/s, MASPs, CD59, Factor B, C3 activation by the alternative pathway, C3

Complement interference in hemorrhagic shock: translation is much needed

Although complement activation has been well described across species to play a crucial role in hemorrhagic shock and subsequent organ damage, to our knowledge, there is so far no real translational approach or clinical trial that investigates therapeutic complement modulation in this setting. In contrast to other injurious insults, in rodent models of hemorrhagic shock (pressure-controlled), complement was robustly activated and led to complement depletion [11], which contributed to

Complement modulation during SIRS: a promising field

It is remarkable that so far no complement modulator has reached approval for clinical use to address sterile posttraumatic SIRS yet. Nevertheless, systemic inflammation during sepsis seems to be a very promising target for the use of complement therapeutics. Interestingly, many compounds utilized in this setting inhibit activation of C3 or C5, highlighting the necessity of a virtually complete block of any downstream pro-inflammatory complement signaling [63], [64], [65]. Inhibition of

Clinical translation and opportunities of complement modulation post trauma

Effective posttraumatic modulation of central complement components (Fig. 1) such as C3, where multiple activation pathways converge, seems reasonable in order to control the fluid phase innate immune response and its intensive cross-talk with other systems. It is somehow surprising that only one single study (the C1-INH compound Cetor® from Sanquin, NCT01275976) so far has addressed the complement system after severe trauma with the results still pending.

On the other hand, interfering with the

Conclusion and prospect

As the main fluid phase branch of innate immunity, complement represents a highly attractive pharmacological target. Very promising results were achieved in preclinical models when complement inhibition was combined with joint inhibition of other targets. In this regard, the dual inhibition of complement and TLR has recently been successfully employed in a porcine model of sepsis [72]. This double inhibition was achieved by co-administration of the tick-derived C5 inhibitor OmCI and an

Financial support

This study was supported by grants from the German Research Foundation DFG (KFO-200) HU 823/3-1,2 and CRC 1149 Project A01 and Z02.

Disclosure

C.Q.S. is an inventor of a patent application that describes the use of miniFH for therapeutic applications. The other authors declare that they have no competing interests as defined by Seminars in Immunology or other interests that might be perceived to influence the results and discussion presented in this paper.

References (82)

  • R. Silasi-Mansat et al.

    Complement inhibition decreases the procoagulant response and confers organ protection in a baboon model of Escherichia coli sepsis

    Blood

    (2010)
  • J.A. Buras et al.

    Inhibition of C5 or absence of C6 protects from sepsis mortality

    Immunobiology

    (2004)
  • D.H. Hangen et al.

    Complement levels in septic primates treated with anti-C5a antibodies

    J. Surg. Res.

    (1989)
  • K. Hoehlig et al.

    A novel C5a-neutralizing mirror-image (l-)aptamer prevents organ failure and improves survival in experimental sepsis

    Mol. Ther.

    (2013)
  • D.C. Mastellos et al.

    Complement-triggered pathways orchestrate regenerative responses throughout phylogenesis

    Semin. Immunol.

    (2013)
  • M.C. Rich et al.

    Site-targeted complement inhibition by a complement receptor 2-conjugated inhibitor (mTT30) ameliorates post-injury neuropathology in mouse brains

    Neurosci. Lett.

    (2016)
  • Z. Fishelson et al.

    Complement and apoptosis

    Mol. Immunol.

    (2001)
  • P. Matzinger

    The danger model: a renewed sense of self

    Science

    (2002)
  • F. Gebhard et al.

    Is interleukin 6 an early marker of injury severity following major trauma in humans?

    Arch. Surg.

    (2000)
  • R.C. Bone, R.A. Balk, F.B. Cerra et al. Definitions for sepsis and organ failure and guidelines for the use of...
  • U. Amara et al.

    Molecular intercommunication between the complement and coagulation systems

    J. Immunol

    (2010)
  • M. Huber-Lang et al.

    Generation of C5a in the absence of C3: a new complement activation pathway

    Nat. Med.

    (2006)
  • F. Gebhard et al.

    Polytrauma–pathophysiology and management principles

    Langenbecks Arch. Surg.

    (2008)
  • K. Balvers et al.

    Risk factors for trauma-induced coagulopathy- and transfusion-associated multiple organ failure in severely injured trauma patients

    Front. Med. (Lausanne)

    (2015)
  • A.M. Burk et al.

    Early complementopathy after multiple injuries in humans

    Shock

    (2012)
  • C. Ehrnthaller et al.

    Alteration of complement hemolytic activity in different trauma and sepsis models

    J. Inflamm. Res.

    (2012)
  • M. Huber-Lang et al.

    Role of complement on broken surfaces after trauma

    Adv. Exp. Med. Biol.

    (2015)
  • Q. Zhang et al.

    Circulating mitochondrial DAMPs cause inflammatory responses to injury

    Nature

    (2010)
  • A.B. Bjornson et al.

    Host defense against opportunist microorganisms following trauma. II. Changes in complement and immunoglobulins in patients with abdominal trauma and in septic patients without trauma

    Ann. Surg.

    (1978)
  • F. Hecke et al.

    Circulating complement proteins in multiple trauma patients–correlation with injury severity, development of sepsis, and outcome

    Crit. Care Med.

    (1997)
  • G. Zilow et al.

    Generation of anaphylatoxin C3a in plasma and bronchoalveolar lavage fluid in trauma patients at risk for the adult respiratory distress syndrome

    Crit. Care Med.

    (1992)
  • T. Muroya et al.

    C4d deposits on the surface of RBCs in trauma patients and interferes with their function

    Crit. Care Med.

    (2014)
  • M.T. Ganter et al.

    Role of the alternative pathway in the early complement activation following major trauma

    Shock

    (2007)
  • L. Lampl et al.

    Hemostatic disorders following polytrauma–the role of physiologic coagulation inhibitors during the preclinical phase

    Anasthesiol. Intensivmed. Notfallmed. Schmerzther.

    (1992)
  • S.M. Kanse et al.

    Factor VII-activating protease is activated in multiple trauma patients and generates anaphylatoxin C5a

    J. Immunol

    (2012)
  • H.I. Kenawy et al.

    Complement-coagulation cross-talk: a potential mediator of the physiological activation of complement by low pH

    Front. Immunol.

    (2015)
  • Z. Fishelson et al.

    Regulation of the alternative pathway of complement by pH

    J. Immunol

    (1987)
  • D. Frith et al.

    Acute traumatic coagulopathy

    Curr. Opin. Anaesthesiol.

    (2012)
  • U. Amara et al.

    Early expression changes of complement regulatory proteins and C5A receptor (CD88) on leukocytes after multiple injury in humans

    Shock

    (2010)
  • J.A. Weigelt et al.

    Complement and the severity of pulmonary failure

    J. Trauma

    (1988)
  • E. Lohde et al.

    Complement activated granulocytes can cause autologous tissue destruction in man

    Mediators Inflamm.

    (1992)
  • Cited by (0)

    View full text