Research paper
Localized delivery of carbon monoxide

https://doi.org/10.1016/j.ejpb.2016.11.002Get rights and content

Abstract

The heme oxygenase (HO)/carbon monoxide (CO) system is a physiological feedback loop orchestrating various cell-protective effects in response to cellular stress. The therapeutic use of CO is impeded by safety challenges as a result of high CO-Hemoglobin formation following non-targeted, systemic administration jeopardizing successful CO therapies as of this biological barrier. Another caveat is the use of CO-Releasing Molecules containing toxicologically critical transition metals. An emerging number of local delivery approaches addressing these issues have recently been introduced and provide exciting new starting points for translating the fascinating preclinical potential of CO into a clinical setting. This review will discuss these approaches and link to future delivery strategies aiming at establishing CO as a safe and effective medication of tomorrow.

Introduction

The heme oxygenase (HO)/carbon monoxide (CO) system is an important cellular feedback-loop towards oxidative and inflammatory insults in man (Fig. 1) [3]. The central building block of this system is the HO-1, an inducible enzyme catalyzing heme degradation into CO, biliverdin, and Fe2+. Detailed reports on cell-protective, anti-oxidative, anti-apoptotic, and anti-inflammatory effects of the HO-1 and its effectors (including CO) have previously been outlined [3]. Polymorphisms impairing upregulation of this enzyme have been linked to various pathophysiological conditions including coronary artery disease [4], diabetes [5], or the outcome of organ transplantations [6]. The disease modifying impact of CO was demonstrated in disease models mainly by systemic application either directly applying CO mostly through inhalation or CO-Releasing Molecules (CORM) [3]. Both modalities face substantial challenges driving the need for novel approaches as discussed below. Inhaled CO leads to one principal challenge of this (and other systemic) administration routes as of the rather exhaustive binding of CO to hemoglobin (Hb), e.g. following diffusion through the alveolar membrane when pulmonary administered. Therefore, systemic trafficking of pulmonary delivered CO is mainly in a tightly bound form as the carboxyhemoglobin (CO-Hb) complex and very little ‘free’ CO is available to act pharmacologically (Fig. 2A). Noteworthy, CO also tightly binds to a variety of other heme-containing proteins including myoglobin [7], neuroglobin [8], and cytochrome C oxidase [9]. For simplification and comparability among various preclinical and clinical reports this review will focus on CO-Hb among these CO binding moieties. The extraordinary stability of CO-Hb requires quite high CO-Hb plasma concentrations such that therapeutically relevant levels of free CO are achieved – critically narrowing the therapeutic window [10]. This conundrum has to be solved for any systemic CO delivery attempt, as very high, perhaps toxic CO-Hb levels have to be condoned in order to achieve therapeutically relevant levels of free CO in the target tissues (Fig. 2A) [11]. This disadvantageous prerequisite is also true for the delivery of CO using systemically administered CORMs. On top, these CORMs aggravate the development risk in that they further hold potentially toxic transition metals required for binding CO (Fig. 2A) [3], [12].

Therefore, and in spite of the exciting therapeutic potential of CO, current systemic application strategies resulted in an uphill battle against both, the CO-Hb to free CO ratio and potential systemic safety concerns associated with the molecular scaffolds most CORMs deploy. Consequently, novel developments point to local delivery and targeting approaches addressing these concerns. These include CORMs that decarbonylate in diseased hepatic tissue (thereby, the CO is carried into the liver before it releases from the CORM – therapeutically relevant free CO is increased while CO-Hb percentages can be kept low) as well as Drug Delivery Systems (DDS) that locally control CO release in the gastrointestinal (GI) tract (thereby further reducing unfavorable CO-Hb binding while maximizing exposure of the target tissue to free CO) [13], [14]. This review will start off past and current CO delivery concepts and therapeutic evidence, define desirable pharmaceutical specifications turning CO into a druggable molecule and finally link to future delivery strategies aiming at translating CO into effective medication of tomorrow.

Nature’s equivalent for the therapeutic approaches discussed here within is HO-1 being central to the local feedback loop of the HO/CO system (Fig. 1). Technologies designed for mimicking this loop, therefore, preferably simulate (i) signaling and (ii) the special spatiotemporal characteristics of this system (vide infra). Resembling the function as a cellular sentinel, the enzyme is induced by an array of diverse imbalances including inflammation, radiation, and hypoxia [15]. Multiple transcriptional factors including activator protein 1 (AP-1), NF-κB, nuclear factor like 2 (NRF-2), cAMP response element-binding protein (CREB), and enhancer box (E-Box) induce HO-1 expression and are themselves controlled through a multitude of pathways including extracellular-signal regulated kinases (ERK), p38 mitogen-activated protein kinases (p38 MAPK), as well as c-Jun N-terminal kinases (JNK) activation [15]. Thereby, HO-1 orchestrates a network of para- and autocrine events providing a protective response (Fig. 1). Autocrine effects are mainly attributed to the high diffusivity and stability of CO, a special characteristic that differentiates CO from other signaling molecules [16]. Hemoglobin is tightly controlling this spatial distribution by instantaneously scavenging CO following passage to the capillary networks (Fig. 1) [17].

The mobility of CO as well as its ability to induce HO-1 has been discussed as important features for effective exogenous application of CO and CORMs [16], [18]. Likewise, genuine HO-1 inducers including cobalt protoporphyrin and hemin have been used as almost equivalent lab stage alternative [3], [19]. Induction is also observed in response to various other compounds including drugs like curcumin [20], acetylsalicylic acid [21], resveratrol [22], lansoprazole [23], [24], and 5-aminosalic acid [25]. The lack of selectivity of this mode of action leading into numerous downstream effects, however, challenged the development of HO-1 inducers for therapeutic purposes [26], [27]. Until today, induction of HO-1 in man was solely demonstrated for hemin [28], which failed in a trial profiling for gastroparesis both in prolonged HO-1 induction (>7 days) and ameliorating the disease activity [29] contrasting previous preclinical trials [30], [31]. Furthermore, hemin is a neurotoxic [32], fairly unselective molecule that besides HO-1 induction represses δ-aminolevulinic acid synthase, the rate-limiting enzyme for the synthesis of tetrapyrrols [15], [33]. This feature is clinically relevant for the treatment of acute porphyria – a rare disease characterized by porphyrin accumulation – for which hemin infusions are used [34]. Therefore, novel effectors of the HO-1 pathways are required and CO is a promising option discussed here within.

Section snippets

Transferability of preclinical results

The systemic application of CO by inhalation demonstrated efficacy in numerous preclinical models [3] contrasting clinical trials: Promising preclinical evidence on endotoxemia [35], [36] was not confirmed in a clinical trial exposing healthy volunteers to 500 ppm CO for one hour resulting in 7% CO-Hb [37]. Likewise, a safety and tolerability study using inhaled CO in kidney transplant patients motivated by promising preclinical results [38], [39], [40] was withdrawn for unknown reason [41].

CO-Releasing Molecules

Metal carbonyl complexes for CO delivery were introduced in 2002 [84]. Starting off these initial studies, various classes of CORMs have been developed with the ultimate goal to combine efficacy and safety as fundamental requirements for drug approvals (Fig. 3). Novel targeting approaches including CORMs for hepatic CO delivery significantly reduce safety issues associated with systemic CO toxicity. Metal toxicity, however, is a golden threat running through all metal based CORMs and has been

Pharmaceutical formulations of CORMs

Within the last years an emerging number of controlled drug delivery systems (DDS) have been introduced for CORMs. Some of these DDS feature first generation CORMS for stimulus driven CO generation, thereby forming an alternative approach to the exciting achievements reported for third generation CORMs (vide supra). Successes were reported for systemic as well as non-invasive use [92], [98].

Conclusion

The therapeutic potential of local CO delivery is enormous as physiological mechanisms have been elucidated and linked to various preclinical mechanistic as well as efficacy studies. Likewise, numerous approaches for the delivery of this challenging gaseous drug have been introduced, thereby providing a versatile toolbox for future translational approaches. Inspired by these, an emerging number of local and target delivery approaches have been introduced addressing the major issue of systemic

References (145)

  • A. Nakao et al.

    Protection against ischemia/reperfusion injury in cardiac and renal transplantation with carbon monoxide, biliverdin and both

    Am. J. Transplant.

    (2005)
  • P. Cabrales et al.

    Hemorrhagic shock resuscitation with carbon monoxide saturated blood

    Resuscitation

    (2007)
  • A. Nakao et al.

    Protective effect of carbon monoxide inhalation for cold-preserved small intestinal grafts

    Surgery

    (2003)
  • E. Beutler

    Effect of carbon-monoxide on red-cell life-span in sickle-cell disease

    Blood

    (1975)
  • D.A. Sears et al.

    Carboxyhemoglobin levels in patients with sickle-cell anemia: relationship to hemolytic and vasoocclusive severity

    Am. J. Med. Sci.

    (2001)
  • C.A. Piantadosi

    Carbon monoxide, reactive oxygen signaling, and oxidative stress

    Free Radic. Biol. Med.

    (2008)
  • L.W. Kao et al.

    Toxicity associated with carbon monoxide

    Clin. Lab. Med.

    (2006)
  • S.S. Mahid et al.

    Smoking and inflammatory bowel disease: a meta-analysis

    Mayo Clin. Proc.

    (2006)
  • W.S. Aronow

    Aggravation of angina pectoris by two percent carboxyhemoglobin

    Am. Heart J.

    (1981)
  • K.S. Davidge et al.

    Carbon monoxide-releasing antibacterial molecules target respiration and global transcriptional regulators

    J. Biol. Chem.

    (2009)
  • G. Saito et al.

    Drug delivery strategy utilizing conjugation via reversible disulfide linkages: role and site of cellular reducing activities

    Adv. Drug Deliv. Rev.

    (2003)
  • X. Ji et al.

    Toward carbon monoxide-based therapeutics: critical drug delivery and developability issues

    J. Pharm. Sci.

    (2016)
  • C. Steiger et al.

    Controlled therapeutic gas delivery systems for quality-improved transplants

    Eur. J. Pharm. Biopharm.

    (2015)
  • H. Yin et al.

    Styrene-maleic acid copolymer-encapsulated CORM2, a water-soluble carbon monoxide (CO) donor with a constant CO-releasing property, exhibits therapeutic potential for inflammatory bowel disease

    J. Control. Release

    (2014)
  • K. Poelstra et al.

    Drug targeting to the diseased liver

    J. Control. Release

    (2012)
  • C.C. Romao et al.

    Developing drug molecules for therapy with carbon monoxide

    Chem. Soc. Rev.

    (2012)
  • N.S. Sitnikov et al.

    Design, synthesis, and functional evaluation of CO-releasing molecules triggered by Penicillin G amidase as a model protease

    Angew. Chem. Int. Ed. Engl.

    (2015)
  • R. Motterlini et al.

    The therapeutic potential of carbon monoxide

    Nat. Rev. Drug Discov.

    (2010)
  • H. Kaneda et al.

    Heme oxygenase-1 gene promoter polymorphism is associated with coronary artery disease in Japanese patients with coronary risk factors

    Arterioscler. Thromb. Vasc. Biol.

    (2002)
  • W. Bao et al.

    Association between heme oxygenase-1 gene promoter polymorphisms and type 2 diabetes mellitus: a HuGE review and meta-analysis

    Am. J. Epidemiol.

    (2010)
  • M. Brunori et al.

    Carbon-monoxide binding by hemoglobin and myoglobin under photodissociating conditions

    Proc. Natl. Acad. Sci. U. S. A.

    (1972)
  • J.L. Wilson et al.

    Design and synthesis of new hybrid molecules that activate the transcription factor Nrf2 and simultaneously release carbon monoxide

    Chemistry (Easton)

    (2014)
  • Q.H. Gibson et al.

    Reactions of cytochrome oxidase with oxygen and carbon monoxide

    Biochem. J.

    (1963)
  • J. Raub

    Carbon Monoxide

    (1999)
  • I.C. Winburn et al.

    Cell damage following carbon monoxide releasing molecule exposure: implications for therapeutic applications

    Basic Clin. Pharmacol. Toxicol.

    (2012)
  • A.R. Marques et al.

    Generation of carbon monoxide releasing molecules (CO-RMs) as drug candidates for the treatment of acute liver injury: targeting of CO-RMs to the liver

    Organometallics

    (2012)
  • A. Prawan et al.

    Molecular basis of heme oxygenase-1 induction: implications for chemoprevention and chemoprotection

    Antioxid. Redox Signal.

    (2005)
  • C.A. Piantadosi

    Biological chemistry of carbon monoxide

    Antioxid. Redox Signal.

    (2002)
  • K.M. Kim et al.

    Carbon monoxide induces heme oxygenase-1 via activation of protein kinase R-like endoplasmic reticulum kinase and inhibits endothelial cell apoptosis triggered by endoplasmic reticulum stress

    Circ. Res.

    (2007)
  • N.G. Abraham et al.

    Pharmacological and clinical aspects of heme oxygenase

    Pharmacol. Rev.

    (2008)
  • Y. Son et al.

    Therapeutic roles of heme oxygenase-1 in metabolic diseases: curcumin and resveratrol analogues as possible inducers of heme oxygenase-1

    Oxid. Med. Cell. Longev.

    (2013)
  • T. Takagi et al.

    Lansoprazole, a proton pump inhibitor, mediates anti-inflammatory effect in gastric mucosal cells through the induction of heme oxygenase-1 via activation of NF-E2-related factor 2 and oxidation of kelch-like ECH-associating protein 1

    J. Pharmacol. Exp. Ther.

    (2009)
  • Y. Yoda et al.

    Prevention by lansoprazole, a proton pump inhibitor, of indomethacin-induced small intestinal ulceration in rats through induction of heme oxygenase-1

    J. Physiol. Pharmacol.

    (2010)
  • A.E. Bharucha et al.

    First-in-human study demonstrating pharmacological activation of heme oxygenase-1 in humans

    Clin. Pharmacol. Ther.

    (2010)
  • A.E. Bharucha et al.

    Effects of hemin on heme oxygenase-1, gastric emptying, and symptoms in diabetic gastroparesis

    Neurogastroenterol. Motil.

    (2016)
  • K.M. Choi et al.

    Heme oxygenase-1 protects interstitial cells of Cajal from oxidative stress and reverses diabetic gastroparesis

    Gastroenterology

    (2008)
  • K.M. Choi et al.

    CD206-positive M2 macrophages that express heme oxygenase-1 protect against diabetic gastroparesis in mice

    Gastroenterology

    (2010)
  • S.R. Robinson et al.

    Hemin toxicity: a preventable source of brain damage following hemorrhagic stroke

    Redox Rep.

    (2009)
  • K.E. Anderson et al.

    Open-label study of hemin for acute porphyria: clinical practice implications

    Am. J. Med.

    (2006)
  • K.E. Anderson et al.

    Recommendations for the diagnosis and treatment of the acute porphyrias

    Ann. Intern. Med.

    (2005)
  • Cited by (0)

    View full text