1
|
Mehrizi TZ, Mirzaei M, Ardestani MS. Pegylation, a Successful Strategy to Address the Storage and Instability Problems of Blood Products: Review 2011-2021. Curr Pharm Biotechnol 2024; 25:247-267. [PMID: 37218184 DOI: 10.2174/1389201024666230522091958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/24/2023] [Accepted: 02/06/2023] [Indexed: 05/24/2023]
Abstract
Conjugation of polyethylene glycol (PEGylation) to blood proteins and cells has emerged as a successful approach to address some of the issues attributed to the storage of blood products, including their short half-life and instability. In this regard, this review study aims to compare the influence of different PEGylation strategies on the quality of several blood products like red blood cells (RBCs), platelets, plasma proteins, i.e., albumin, coagulation factor VIII, and antibodies. The results indicated that conjugating succinimidyl carbonate methoxyPEG (SCmPEG) to platelets could improve blood transfusion safety by preventing these cells from being attached to low-load hidden bacteria in blood products. Moreover, coating of 20 kD succinimidyl valerate (SVA)-mPEG to RBCs was able to extend the half-life and stability of these cells during storage, as well as immune camouflage their surface antigens to prevent alloimmunisation. As regards albumin products, PEGylation improved the albumin stability, especially during sterilization, and there was a relationship between the molecular weight (MW) of PEG molecules and the biological half-life of the conjugate. Although coating antibodies with short-chain PEG molecules could enhance their stabilities, these modified proteins were cleared from the blood faster. Also, branched PEG molecules enhanced the retention and shielding of the fragmented and bispecific antibodies. Overall, the results of this literature review indicate that PEGylation can be considered a useful tool for enhancing the stability and storage of blood components.
Collapse
Affiliation(s)
| | - Mehdi Mirzaei
- Iran Ministry of Health and Medical Education, Deputy Ministry for Education, Tehran, Iran
| | - Mehdi Shafiee Ardestani
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Arrigo E, Comità S, Pagliaro P, Penna C, Mancardi D. Clinical Applications for Gasotransmitters in the Cardiovascular System: Are We There Yet? Int J Mol Sci 2023; 24:12480. [PMID: 37569855 PMCID: PMC10419417 DOI: 10.3390/ijms241512480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Ischemia is the underlying mechanism in a wide variety of acute and persistent pathologies. As such, understanding the fine intracellular events occurring during (and after) the restriction of blood supply is pivotal to improving the outcomes in clinical settings. Among others, gaseous signaling molecules constitutively produced by mammalian cells (gasotransmitters) have been shown to be of potential interest for clinical treatment of ischemia/reperfusion injury. Nitric oxide (NO and its sibling, HNO), hydrogen sulfide (H2S), and carbon monoxide (CO) have long been proven to be cytoprotective in basic science experiments, and they are now awaiting confirmation with clinical trials. The aim of this work is to review the literature and the clinical trials database to address the state of development of potential therapeutic applications for NO, H2S, and CO and the clinical scenarios where they are more promising.
Collapse
|
3
|
Russo I, Barale C, Melchionda E, Penna C, Pagliaro P. Platelets and Cardioprotection: The Role of Nitric Oxide and Carbon Oxide. Int J Mol Sci 2023; 24:ijms24076107. [PMID: 37047079 PMCID: PMC10094148 DOI: 10.3390/ijms24076107] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Nitric oxide (NO) and carbon monoxide (CO) represent a pair of biologically active gases with an increasingly well-defined range of effects on circulating platelets. These gases interact with platelets and cells in the vessels and heart and exert fundamentally similar biological effects, albeit through different mechanisms and with some peculiarity. Within the cardiovascular system, for example, the gases are predominantly vasodilators and exert antiaggregatory effects, and are protective against damage in myocardial ischemia-reperfusion injury. Indeed, NO is an important vasodilator acting on vascular smooth muscle and is able to inhibit platelet activation. NO reacts with superoxide anion (O2(-•)) to form peroxynitrite (ONOO(-)), a nitrosating agent capable of inducing oxidative/nitrative signaling and stress both at cardiovascular, platelet, and plasma levels. CO reduces platelet reactivity, therefore it is an anticoagulant, but it also has some cardioprotective and procoagulant properties. This review article summarizes current knowledge on the platelets and roles of gas mediators (NO, and CO) in cardioprotection. In particular, we aim to examine the link and interactions between platelets, NO, and CO and cardioprotective pathways.
Collapse
Affiliation(s)
- Isabella Russo
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Cristina Barale
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Elena Melchionda
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences of Turin University, Orbassano, I-10043 Turin, Italy
| |
Collapse
|
4
|
Holz E, Darwish M, Tesar DB, Shatz-Binder W. A Review of Protein- and Peptide-Based Chemical Conjugates: Past, Present, and Future. Pharmaceutics 2023; 15:600. [PMID: 36839922 PMCID: PMC9959917 DOI: 10.3390/pharmaceutics15020600] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Over the past few decades, the complexity of molecular entities being advanced for therapeutic purposes has continued to evolve. A main propellent fueling innovation is the perpetual mandate within the pharmaceutical industry to meet the needs of novel disease areas and/or delivery challenges. As new mechanisms of action are uncovered, and as our understanding of existing mechanisms grows, the properties that are required and/or leveraged to enable therapeutic development continue to expand. One rapidly evolving area of interest is that of chemically enhanced peptide and protein therapeutics. While a variety of conjugate molecules such as antibody-drug conjugates, peptide/protein-PEG conjugates, and protein conjugate vaccines are already well established, others, such as antibody-oligonucleotide conjugates and peptide/protein conjugates using non-PEG polymers, are newer to clinical development. This review will evaluate the current development landscape of protein-based chemical conjugates with special attention to considerations such as modulation of pharmacokinetics, safety/tolerability, and entry into difficult to access targets, as well as bioavailability. Furthermore, for the purpose of this review, the types of molecules discussed are divided into two categories: (1) therapeutics that are enhanced by protein or peptide bioconjugation, and (2) protein and peptide therapeutics that require chemical modifications. Overall, the breadth of novel peptide- or protein-based therapeutics moving through the pipeline each year supports a path forward for the pursuit of even more complex therapeutic strategies.
Collapse
Affiliation(s)
- Emily Holz
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Martine Darwish
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Devin B. Tesar
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Whitney Shatz-Binder
- Department of Pharmaceutical Development, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
- Department of Protein Chemistry, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
5
|
Scalable production and complete biophysical characterization of poly(ethylene glycol) surface conjugated liposome encapsulated hemoglobin (PEG-LEH). PLoS One 2022; 17:e0269939. [PMID: 35802716 PMCID: PMC9269976 DOI: 10.1371/journal.pone.0269939] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/31/2022] [Indexed: 11/19/2022] Open
Abstract
Particle encapsulated hemoglobin (Hb)-based oxygen (O2) carriers (HBOCs) have clear advantages over their acellular counterparts because of their larger molecular diameter and lack of vasoactivity upon transfusion. Poly(ethylene glycol) surface conjugated liposome encapsulated Hb (PEG-LEH) nanoparticles are considered a promising class of HBOC for use as a red blood cell (RBC) substitute. However, their widespread usage is limited by manufacturing processes which prevent material scale up. In this study, PEG-LEH nanoparticles were produced via a scalable and robust process using a high-pressure cell disruptor, and their biophysical properties were thoroughly characterized. Hb encapsulation, methemoglobin (metHb) level, O2-PEG-LEH equilibria, PEG-LEH gaseous (oxygen, carbon monoxide, nitric oxide) ligand binding/release kinetics, lipocrit, and long-term storage stability allowed us to examine their potential suitability and efficacy as an RBC replacement. Our results demonstrate that PEG-LEH nanoparticle suspensions manufactured via a high-pressure cell disruptor have Hb concentrations comparable to whole blood (~12 g/dL) and possess other desirable characteristics, which may permit their use as potential lifesaving O2 therapeutics.
Collapse
|
6
|
Charbe NB, Castillo F, Tambuwala MM, Prasher P, Chellappan DK, Carreño A, Satija S, Singh SK, Gulati M, Dua K, González-Aramundiz JV, Zacconi FC. A new era in oxygen therapeutics? From perfluorocarbon systems to haemoglobin-based oxygen carriers. Blood Rev 2022; 54:100927. [PMID: 35094845 DOI: 10.1016/j.blre.2022.100927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/14/2021] [Accepted: 01/12/2022] [Indexed: 02/09/2023]
|
7
|
Galili U, Zhu Z, Chen J, Goldufsky JW, Schaer GL. Near Complete Repair After Myocardial Infarction in Adult Mice by Altering the Inflammatory Response With Intramyocardial Injection of α-Gal Nanoparticles. Front Cardiovasc Med 2021; 8:719160. [PMID: 34513957 PMCID: PMC8425953 DOI: 10.3389/fcvm.2021.719160] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/30/2021] [Indexed: 12/27/2022] Open
Abstract
Background: Neonatal mice, but not older mice, can regenerate their hearts after myocardial-infarction (MI), a process mediated by pro-reparative macrophages. α-Gal nanoparticles applied to skin wounds in adult-mice bind the anti-Gal antibody, activate the complement cascade and generate complement chemotactic peptides that recruit pro-reparative macrophages which are further activated by these nanoparticles. The recruited macrophages decrease wound healing time by ~50%, restore the normal skin structure and prevent fibrosis and scar formation in mice. Objectives: The objective of this study is to determine if α-gal nanoparticles injected into the reperfused myocardium after MI in adult-mice can induce myocardial repair that restores normal structure, similar to that observed in skin injuries. Methods and Results: MI was induced by occluding the mid-portion of the left anterior descending (LAD) coronary artery for 30 min. Immediately following reperfusion, each mouse received two 10 μl injections of 100 μg α-gal nanoparticles in saline into the LAD territory (n = 20), or saline for controls (n = 10). Myocardial infarct size was measured by planimetry following Trichrome staining and macrophage recruitment by hematoxylin-eosin staining. Left ventricular (LV) function was measured by echocardiography. Control mice displayed peak macrophage infiltration at 4-days, whereas treated mice had a delayed peak macrophage infiltration at 7-days. At 28-days, control mice demonstrated large transmural infarcts with extensive scar formation and poor contractile function. In contrast, mice treated with α-gal nanoparticles demonstrated after 28-days a marked reduction in infarct size (~10-fold smaller), restoration of normal myocardium structure and contractile function. Conclusions: Intramyocardial injection of α-gal nanoparticles post-MI in anti-Gal producing adult-mice results in near complete repair of the infarcted territory, with restoration of normal LV structure and contractile function. The mechanism responsible for this benefit likely involves alteration of the usual inflammatory response post-MI, as previously observed with regeneration of injured hearts in adult zebrafish, salamanders and neonatal mice.
Collapse
Affiliation(s)
- Uri Galili
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Zhongkai Zhu
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Jiwang Chen
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Josef W Goldufsky
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Gary L Schaer
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
8
|
Microvascular and Systemic Impact of Resuscitation with PEGylated Carboxyhemoglobin-Based Oxygen Carrier or Hetastarch in a Rat Model of Transient Hemorrhagic Shock. Shock 2021; 53:493-502. [PMID: 31045989 DOI: 10.1097/shk.0000000000001370] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Hemorrhage is the leading cause of preventable, traumatic death. Currently, prehospital resuscitation fluids provide preload but not oxygen-carrying capacity-a critical blood function that mitigates microvascular ischemia and tissue hypoxia during hemorrhagic shock. Solutions containing polymerized hemoglobin have been associated with vasoactive and hypertensive events. A novel hemoglobin-based oxygen carrier, modified with PEGylation and CO moieties (PEG-COHb), may overcome these limitations. OBJECTIVES To evaluate the systemic and microcirculatory effects of PEG-COHb as compared with the 6% hetastarch in a rat model of hemorrhagic shock. METHODS Male Sprague Dawley rats (N = 20) were subjected to severe, controlled, hemorrhagic shock. Animals were randomized to 20% estimated blood-volume resuscitation with either 6% hetastarch or PEG-COHb. Continuous, invasive, cardiovascular measurements, and arterial blood gases were measured. Microcirculatory measurements of interstitial oxygenation (PISFO2) and vasoactivity helped model oxygen delivery in the spinotrapezius muscle using intravital and phosphorescence quenching microscopy. RESULTS Hemorrhage reduced mean arterial pressure (MAP), arteriolar diameter, and PISFO2, and increased lactate 10-fold in both groups. Resuscitation with both PEG-COHb and hetastarch improved cardiovascular parameters. However, PEG-COHb treatment resulted in higher MAP (P < 0.001), improved PISFO2 (14 [PEG-COHb] vs. 5 [hetastarch] mmHg; P < 0.0001), lower lactate post-resuscitation (P < 0.01), and extended survival from 90 to 142 min (P < 0.001) as compared with the hetastarch group. CONCLUSIONS PEG-COHb improved MAP PISFO2, lactate, and survival time as compared with 6% hetastarch resuscitation. Importantly, hypertension and vasoactivity were not detected in response to PEG-COHb resuscitation supporting further investigation of this resuscitation strategy.
Collapse
|
9
|
Macko A, Sheppard FR, Nugent WH, Abuchowski A, Song BK. Improved Hemodynamic Recovery and 72-Hour Survival Following Low-Volume Resuscitation with a PEGylated Carboxyhemoglobin in a Rat Model of Severe Hemorrhagic Shock. Mil Med 2021; 185:e1065-e1072. [PMID: 32302002 DOI: 10.1093/milmed/usz472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/15/2019] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Hemorrhage is a leading cause of death from potentially survivable civilian and military trauma. As projected conflicts move from settings of tactical and logistical supremacy to hyper-dynamic tactical zones against peer and near-peer adversaries, protracted medical evacuation times are expected. Treatment at the point-of-injury is critical. Although crystalloids like Lactated Ringer's (LR) are ubiquitous, whole blood (WB) is the preferred resuscitation fluid following hemorrhage; however, logistical constraints limit the availability of WB in prehospital settings. Hemoglobin-based oxygen carriers (HBOCs) offer both hemodynamic support and oxygen-carrying capacity while avoiding logistical constraints of WB. We hypothesized that low-volume resuscitation of severe hemorrhagic shock with an HBOC (PEGylated carboxyhemoglobin, [PC]) would improve hemodynamic recovery and 72-hour survival; comparable to WB and superior to LR. MATERIALS AND METHODS A total of 21 anesthetized male Sprague-Dawley rats underwent severe hemorrhagic shock followed by randomly assigned low-volume resuscitation with LR, WB, or PC, and then recovered from anesthesia for up to 72-hour observation. Mean arterial pressure (MAP) was recorded continuously under anesthesia, and arterial blood gases were measured at baseline (BL), 60 minutes post-hemorrhage (HS1h), and 24 hours post-resuscitation (PR24h). Survival was presented on a Kaplan-Meier plot and significance determined with a log-rank test. Cardiovascular and blood gas data were assessed with one-way analysis of variance and post hoc analysis where appropriate. RESULTS All measured cardiovascular and blood chemistry parameters were equivalent between groups at BL and HS1h. BL MAP values were 90 ± 3, 86 ± 1, and 89 ± 2 mmHg for LR, PC, and WB, respectively. Immediately following resuscitation, MAP values were 57 ± 4, 74 ± 5, and 62 ± 3 mmHg, with PC equivalent to WB and higher than LR (P < 0.05). WB and LR were both lower than BL (P < 0.0001), whereas PC was not (P = 0.13). The PC group's survival to 72 hours was 57%, which was not different from WB (43%) and higher than LR (14%; P < 0.05). CONCLUSIONS A single bolus infusion of PC produced superior survival and MAP response compared to LR, which is the standard fluid resuscitant carried by combat medics. PC was not different from WB in terms of survival and MAP, which is encouraging because its reduced logistical constraints make it viable for field deployment. These promising findings warrant further development and investigation of PC as a low-volume, early treatment for hemorrhagic shock in scenarios where blood products may not be available.
Collapse
Affiliation(s)
- Antoni Macko
- Song Biotechnologies, 855 N Wolfe St., Suite 622, Baltimore, MD 21205 USA
| | - Forest R Sheppard
- Department of Surgery, Division of Acute Care Surgery, Maine Medical Center, 887 Congress St #400, Portland, ME 04102
| | - William H Nugent
- Song Biotechnologies, 855 N Wolfe St., Suite 622, Baltimore, MD 21205 USA
| | - Abe Abuchowski
- Prolong Pharmaceuticals, 300 Corporate Ct, South Plainfield, NJ 07080
| | - Bjorn K Song
- Song Biotechnologies, 855 N Wolfe St., Suite 622, Baltimore, MD 21205 USA
| |
Collapse
|
10
|
Muller CR, Williams AT, Munoz CJ, Eaker AM, Breton AN, Palmer AF, Cabrales P. Safety profile of high molecular weight polymerized hemoglobins. Transfusion 2020; 61:212-224. [PMID: 33104250 DOI: 10.1111/trf.16157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hemoglobin (Hb)-based oxygen (O2 ) carriers (HBOCs) are being developed as alternatives to red blood cells and blood when these products are unavailable. Clinical trials of previous HBOC generations revealed side effects, including hypertension and vasoconstriction, that were not observed in preclinical studies. Large molecular weight (MW) polymerized bovine Hb (PolybHb) represents a new class of HBOC with promising results. We evaluated the safety profile of PolybHb after an exchange transfusion (ET) in guinea pigs (GPs). This study compares changes in indices of cardiac, inflammatory, and organ function after ET with high (R-state) and low (T-state) O2 affinity PolybHb with high MW. STUDY DESIGN AND METHODS Guinea pigs underwent a 20% ET with PolybHb. To assess the implication of PolybHb ET on the microcirculation, hamsters instrumented with a dorsal window chamber were subjected to a similar volume ET. RESULTS T and R-state PolybHb did not induce significant alterations in cardiac function. T-state PolybHb induced mild vasoconstriction shortly after transfusion, while R-state did not have acute effects on microvascular tone. CONCLUSION Large MW PolybHbs were found to be safe and efficacious in increasing O2 carrying capacity and the O2 affinity of the PolybHb did not affect O2 delivery or extraction by tissues in relevant preclinical models. In conclusion, these results suggest that both T-state and R-state PolybHb are safe and do not impair O2 delivery. The results are encouraging and support further evaluation of high MW PolybHbs and their future feasibility compared to allogenic blood in a trauma model.
Collapse
Affiliation(s)
- Cynthia R Muller
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Alexander T Williams
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Carlos J Munoz
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Allyn M Eaker
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Amanda N Breton
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Andre F Palmer
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Pedro Cabrales
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
11
|
Taguchi K, Maruyama T, Otagiri M. Use of Hemoglobin for Delivering Exogenous Carbon Monoxide in Medicinal Applications. Curr Med Chem 2020; 27:2949-2963. [PMID: 30421669 DOI: 10.2174/0929867325666181113122340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/25/2018] [Accepted: 11/09/2018] [Indexed: 01/02/2023]
Abstract
Carbon Monoxide (CO), at low concentrations, can have a variety of positive effects on the body including anti-apoptosis, anti-inflammatory, anti-oxidative and anti-proliferative effects. Although CO has great potential for use as a potent medical bioactive gas, for it to exist in the body in stable form, it must be associated with a carrier. Hemoglobin (Hb) represents a promising material for use as a CO carrier because most of the total CO in the body is stored associated with Hb in red blood cells (RBC). Attempts have been made to develop an Hb-based CO carrying system using RBC and Hb-based artificial oxygen carriers. Some of these have been reported to be safe and to have therapeutic value as a CO donor in preclinical and clinical studies. In the present review, we overview the potential of RBC and Hb-based artificial oxygen carriers as CO carriers based on the currently available literature evidence for their use in pharmaceutical therapy against intractable disorders.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan.,DDS Research Institute, Sojo University, Kumamoto, Japan
| |
Collapse
|
12
|
Abstract
Supplemental Digital Content is available in the text Background: Hemorrhage and its complications are the leading cause of preventable death from trauma in young adults, especially in remote locations. To address this, deliverable, shelf-stable resuscitants that provide therapeutic benefits throughout the time course of hemorrhagic shock and the progressive ischemic injury it produces are needed. SANGUINATE∗ is a novel bovine PEGylated carboxyhemoglobin-based oxygen carrier, which has desirable oxygen-carrying and oncotic properties as well as a CO moiety to maintain microvascular perfusion. Objectives: To compare the crystalloid (Lactated Ringer's Solution; LRS), and the colloid (Hextend†) standards of care with SANGUINATE in a post “golden hour” resuscitation model. Methods: Rats underwent a controlled, stepwise blood withdrawal (45% by volume), were maintained in untreated hemorrhagic shock state for >60 min, resuscitated with a 20% bolus of one of the three test solutions, and observed till demise. Parameters of tissue oxygenation (PISFO2), arteriolar diameters, and mean arterial pressure (MAP) were collected. Results: SANGUINATE-treated animals survived significantly longer than those treated with Hextend and LRS. SANGUINATE also significantly increased tissue PISFO2 2 h after resuscitation, whereas LRS and Hextend did not. SANGUINATE also produced a significantly higher MAP, which was hypotensive compared to baseline, that endured until demise. Conclusions: Resuscitation with SANGUINATE after prolonged hemorrhagic shock improves survival, MAP, and PISFO2 compared with standard of care plasma expanders. Since the pathologies of hemorrhagic shock and the associated systemic ischemia are progressive, preclinical studies of this nature are essential to determine efficacy of new resuscitants across the range of possible times to treatment.
Collapse
|
13
|
Abstract
In blood, the primary role of red blood cells (RBCs) is to transport oxygen via highly regulated mechanisms involving hemoglobin (Hb). Hb is a tetrameric porphyrin protein comprising of two α- and two β-polypeptide chains, each containing an iron-containing heme group capable of binding one oxygen molecule. In military as well as civilian traumatic exsanguinating hemorrhage, rapid loss of RBCs can lead to suboptimal tissue oxygenation and subsequent morbidity and mortality. In such cases, transfusion of whole blood or RBCs can significantly improve survival. However, blood products including RBCs present issues of limited availability and portability, need for type matching, pathogenic contamination risks, and short shelf-life, causing substantial logistical barriers to their prehospital use in austere battlefield and remote civilian conditions. While robust research is being directed to resolve these issues, parallel research efforts have emerged toward bioengineering of semisynthetic and synthetic surrogates of RBCs, using various cross-linked, polymeric, and encapsulated forms of Hb. These Hb-based oxygen carriers (HBOCs) can potentially provide therapeutic oxygenation when blood or RBCs are not available. Several of these HBOCs have undergone rigorous preclinical and clinical evaluation, but have not yet received clinical approval in the USA for human use. While these designs are being optimized for clinical translations, several new HBOC designs and molecules have been reported in recent years, with unique properties. The current article will provide a comprehensive review of such HBOC designs, including current state-of-the-art and novel molecules in development, along with a critical discussion of successes and challenges in this field.
Collapse
|
14
|
Effect of Polyethylene-glycolated Carboxyhemoglobin on Renal Microcirculation in a Rat Model of Hemorrhagic Shock. Anesthesiology 2020; 131:1110-1124. [PMID: 31490291 DOI: 10.1097/aln.0000000000002932] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Primary resuscitation fluid to treat hemorrhagic shock remains controversial. Use of hydroxyethyl starches raised concerns of acute kidney injury. Polyethylene-glycolated carboxyhemoglobin, which has carbon monoxide-releasing molecules and oxygen-carrying properties, was hypothesized to sustain cortical renal microcirculatory PO2 after hemorrhagic shock and reduce kidney injury. METHODS Anesthetized and ventilated rats (n = 42) were subjected to pressure-controlled hemorrhagic shock for 1 h. Renal cortical PO2 was measured in exposed kidneys using a phosphorescence quenching method. Rats were randomly assigned to six groups: polyethylene-glycolated carboxyhemoglobin 320 mg · kg, 6% hydroxyethyl starch (130/0.4) in Ringer's acetate, blood retransfusion, diluted blood retransfusion (~4 g · dl), nonresuscitated animals, and time control. Nitric oxide and heme oxygenase 1 levels were determined in plasma. Kidney immunohistochemistry (histologic scores of neutrophil gelatinase-associated lipocalin and tumor necrosis factor-α) and tubular histologic damages analyses were performed. RESULTS Blood and diluted blood restored renal PO2 to 51 ± 5 mmHg (mean difference, -18; 95% CI, -26 to -11; P < 0.0001) and 47 ± 5 mmHg (mean difference, -23; 95% CI, -31 to -15; P < 0.0001), respectively, compared with 29 ± 8 mmHg for hydroxyethyl starch. No differences between polyethylene-glycolated carboxyhemoglobin and hydroxyethyl starch were observed (33 ± 7 mmHg vs. 29 ± 8 mmHg; mean difference, -5; 95% CI, -12 to 3; P = 0.387), but significantly less volume was administered (4.5 [3.3-6.2] vs. 8.5[7.7-11.4] ml; mean rank difference, 11.98; P = 0.387). Blood and diluted blood increased the plasma bioavailability of nitric oxide compared with hydroxyethyl starch (mean rank difference, -20.97; P = 0.004; and -17.13; P = 0.029, respectively). No changes in heme oxygenase 1 levels were observed. Polyethylene-glycolated carboxyhemoglobin limited tubular histologic damages compared with hydroxyethyl starch (mean rank difference, 60.12; P = 0.0012) with reduced neutrophil gelatinase-associated lipocalin (mean rank difference, 84.43; P < 0.0001) and tumor necrosis factor-α (mean rank difference, 49.67; P = 0.026) histologic scores. CONCLUSIONS Polyethylene-glycolated carboxyhemoglobin resuscitation did not improve renal PO2 but limited tubular histologic damages and neutrophil gelatinase-associated lipocalin upregulation after hemorrhage compared with hydroxyethyl starch, whereas a lower volume was required to sustain macrocirculation.
Collapse
|
15
|
Khan F, Singh K, Friedman MT. Artificial Blood: The History and Current Perspectives of Blood Substitutes. Discoveries (Craiova) 2020; 8:e104. [PMID: 32309621 PMCID: PMC7086064 DOI: 10.15190/d.2020.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/07/2020] [Accepted: 03/07/2020] [Indexed: 01/09/2023] Open
Abstract
Blood transfusions are one of the most common procedures performed in hospitalized patients. Yet, despite all of the measures taken to ensure the safety of the blood supply, there are known risks associated with transfusions, including infectious and noninfectious complications. Meanwhile, issues with blood product availability, the need for compatibility testing, and the storage and transport requirements of blood products, have presented challenges for the administration of blood transfusions. Additionally, there are individuals who do not accept blood transfusions (e.g., Jehovah's Witnesses). Therefore, there is a need to develop alternative agents that can reliably and safely replace blood. However, although there have been many attempts to develop blood substitutes over the years, there are currently no such products available that have been approved by the United States Food and Drug Administration (FDA). However, a more-recently developed hemoglobin-based oxygen carrier has shown promise in early clinical trials and has achieved the status of "Orphan Drug" under the FDA.
Collapse
Affiliation(s)
- Fahad Khan
- Mount Sinai Health System, Department of Pathology and Laboratory Medicine, Icahn School of Medicine, New York, NY, USA
| | - Kunwar Singh
- Mount Sinai Health System, Department of Pathology and Laboratory Medicine, Icahn School of Medicine, New York, NY, USA
| | - Mark T. Friedman
- Mount Sinai Health System, Department of Pathology and Laboratory Medicine, Icahn School of Medicine, New York, NY, USA
| |
Collapse
|
16
|
Kawaguchi AT, Salybekov AA, Yamano M, Kitagishi H, Sekine K, Tamaki T. PEGylated carboxyhemoglobin bovine (SANGUINATE) ameliorates myocardial infarction in a rat model. Artif Organs 2019; 42:1174-1184. [PMID: 30375680 PMCID: PMC6590322 DOI: 10.1111/aor.13384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/28/2018] [Accepted: 10/22/2018] [Indexed: 12/24/2022]
Abstract
Artificial oxygen (O2) carriers were reported to be protective in ischemia/reperfusion (I/R) in various organs including the heart. In the current study, 20 rats underwent ligation (MI) of the left anterior descending artery, were treated with 10 mL/kg of PEGylated carboxyhemoglobin bovine (SANGUINATE, S+, n = 10) or saline (S−, n = 10) 10 minutes after MI and daily thereafter for 3 days, and were followed by weekly echocardiography for 4 weeks, when they had left ventricular pressure volume relationship (PVR) analyses followed by necropsy. Echocardiography showed an increase in end‐systolic dimension rather than end‐diastolic dimension, preserved fractional shortening (36 vs. 26%, P < .01), and milder mitral regurgitation in S+ compared with S‐ rats. PVR revealed a milder increase in end‐systolic volume, larger stroke volume (101 vs. 74 μL, P < .005) and cardiac output (33.4 vs. 23.8 mL/min, P = .004) in S+ rats in actual determination and under a wide range of standardized loading conditions 4 weeks after MI. Excised heart showed significantly limited area of MI (8.9 vs. 13.3%, P = .028). The results suggest that SANGUINATE in short‐term repeated doses may accelerate weight recovery, preserving the myocardium, mitral competence, and cardiac function after MI. The mechanism of action and optimal treatment for MI remain to be studied.
Collapse
Affiliation(s)
- Akira T Kawaguchi
- Cell Transplantation and Regenerative Medicine, Tokai University, Kanagawa, Japan
| | | | - Mariko Yamano
- Department of Rehabilitation, Osaka Prefecture University, Osaka, Japan
| | - Hiroaki Kitagishi
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyoto, Japan
| | - Kaori Sekine
- Department of Human Structure and Function, Tokai University, Kanagawa, Japan
| | - Tetsuro Tamaki
- Department of Human Structure and Function, Tokai University, Kanagawa, Japan
| |
Collapse
|
17
|
Nugent WH, Jubin R, Buontempo PJ, Kazo F, Song BK. Microvascular and systemic responses to novel PEGylated carboxyhaemoglobin-based oxygen carrier in a rat model of vaso-occlusive crisis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:95-103. [DOI: 10.1080/21691401.2018.1543197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
| | - Ronald Jubin
- Prolong Pharmaceuticals, South Plainfield, NJ, USA
| | | | | | | |
Collapse
|
18
|
Meng F, Kaul D, Thangaswamy S, Bhutoria S, Gerfen G, Branch C, Intaglietta M, Acharya SA. Semisynthetic supra plasma expanders: a new class of therapeutics to improve microcircualtion in sickle cell anaemia. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:73-82. [DOI: 10.1080/21691401.2018.1543194] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Fantao Meng
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MA, USA
| | - Dhananjaya Kaul
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Savita Bhutoria
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gary Gerfen
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Craig Branch
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Marcos Intaglietta
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Seetharama A. Acharya
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
19
|
DeSimone RA, Berlin DA, Avecilla ST, Goss CA. Investigational use of PEGylated carboxyhemoglobin bovine in a Jehovah's Witness with hemorrhagic shock. Transfusion 2018; 58:2297-2300. [PMID: 30203845 DOI: 10.1111/trf.14799] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/29/2018] [Accepted: 04/19/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Jehovah's Witnesses pose a clinical challenge in the setting of critical anemia. Most do not accept transfusions, but some accept hemoglobin-based oxygen carriers on a compassionate-use basis. PEGylated carboxyhemoglobin bovine (PCHB) is an acellular dual-action carbon monoxide (CO)-releasing and oxygen transfer agent currently being investigated in Phase II clinical trials. CASE REPORT We present the case of a 42-year-old Jehovah's Witness with an acute upper gastrointestinal bleed and hemorrhagic shock who required emergent PCHB for stabilization during lifesaving interventions. After PCHB infusion, the patient's shock and encephalopathy improved with decreased vasopressor requirement. Through gastroenterology and interventional radiology procedures, the patient's bleeding stabilized. While receiving five additional doses of PCHB and other supportive therapies (iron, folate, vitamin B12, darbepoetin alfa), the patient was extubated and weaned off vasopressors. CONCLUSIONS PCHB was used to stabilize (bridge) a critically ill anemic patient for lifesaving interventions without adverse effects. Additional studies are warranted to explore the drug's safety profile and efficacy in patients declining blood products.
Collapse
Affiliation(s)
| | - David A Berlin
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York-Presbyterian Hospital-Weill Cornell Medicine
| | - Scott T Avecilla
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cheryl A Goss
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
20
|
Thiagarajan D, O’ Shea K, Sreejit G, Ananthakrishnan R, Quadri N, Li Q, Schmidt AM, Gabbay K, Ramasamy R. Aldose reductase modulates acute activation of mesenchymal markers via the β-catenin pathway during cardiac ischemia-reperfusion. PLoS One 2017; 12:e0188981. [PMID: 29190815 PMCID: PMC5708684 DOI: 10.1371/journal.pone.0188981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022] Open
Abstract
Aldose reductase (AR: human, AKR1B1; mouse, AKR1B3), the first enzyme in the polyol pathway, plays a key role in mediating myocardial ischemia/reperfusion (I/R) injury. In earlier studies, using transgenic mice broadly expressing human AKR1B1 to human-relevant levels, mice devoid of Akr1b3, and pharmacological inhibitors of AR, we demonstrated that AR is an important component of myocardial I/R injury and that inhibition of this enzyme protects the heart from I/R injury. In this study, our objective was to investigate if AR modulates the β-catenin pathway and consequent activation of mesenchymal markers during I/R in the heart. To test this premise, we used two different experimental models: in vivo, Akr1b3 null mice and wild type C57BL/6 mice (WT) were exposed to acute occlusion of the left anterior descending coronary artery (LAD) followed by recovery for 48 hours or 28 days, and ex-vivo, WT and Akr1b3 null murine hearts were perfused using the Langendorff technique (LT) and subjected to 30 min of global (zero-flow) ischemia followed by 60 min of reperfusion. Our in vivo results reveal reduced infarct size and improved functional recovery at 48 hours in mice devoid of Akr1b3 compared to WT mice. We demonstrate that the cardioprotection observed in Akr1b3 null mice was linked to acute activation of the β-catenin pathway and consequent activation of mesenchymal markers and genes linked to fibrotic remodeling. The increased activity of the β-catenin pathway at 48 hours of recovery post-LAD was not observed at 28 days post-infarction, thus indicating that the observed increase in β-catenin activity was transient in the mice hearts devoid of Akr1b3. In ex vivo studies, inhibition of β-catenin blocked the cardioprotection observed in Akr1b3 null mice hearts. Taken together, these data indicate that AR suppresses acute activation of β-catenin and, thereby, blocks consequent induction of mesenchymal markers during early reperfusion after myocardial ischemia. Inhibition of AR might provide a therapeutic opportunity to optimize cardiac remodeling after I/R injury.
Collapse
Affiliation(s)
- Devi Thiagarajan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Karen O’ Shea
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Gopalkrishna Sreejit
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Radha Ananthakrishnan
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Nosirudeen Quadri
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Qing Li
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Ann Marie Schmidt
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
| | - Kenneth Gabbay
- Department of Pediatrics, Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ravichandran Ramasamy
- Diabetes Research Program, Department of Medicine, New York University Langone Medical Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Dhar R, Misra H, Diringer MN. SANGUINATE™ (PEGylated Carboxyhemoglobin Bovine) Improves Cerebral Blood Flow to Vulnerable Brain Regions at Risk of Delayed Cerebral Ischemia After Subarachnoid Hemorrhage. Neurocrit Care 2017. [DOI: 10.1007/s12028-017-0418-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Sam C, Desai P, Laber D, Patel A, Visweshwar N, Jaglal M. Pegylated bovine carboxyhaemoglobin utilisation in a thrombotic thrombocytopenic purpura patient. Transfus Med 2017; 27:300-302. [DOI: 10.1111/tme.12407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 12/14/2022]
Affiliation(s)
- C. Sam
- Department of Hematology; University of South Florida Morsani College of Medicine; Tampa Florida USA
| | - P. Desai
- Department of Hematology; University of South Florida Morsani College of Medicine; Tampa Florida USA
| | - D. Laber
- Department of Hematology; University of South Florida Morsani College of Medicine; Tampa Florida USA
- Department of Hematology & Oncology; Moffitt Cancer Center; Tampa Florida USA
| | - A. Patel
- Department of Hematology; University of South Florida Morsani College of Medicine; Tampa Florida USA
- Department of Hematology & Oncology; Moffitt Cancer Center; Tampa Florida USA
| | - N. Visweshwar
- Department of Hematology; University of South Florida Morsani College of Medicine; Tampa Florida USA
| | - M. Jaglal
- Department of Hematology; University of South Florida Morsani College of Medicine; Tampa Florida USA
- Department of Hematology & Oncology; Moffitt Cancer Center; Tampa Florida USA
| |
Collapse
|
23
|
Webster KD, Dahhan D, Otto AM, Frosti CL, Dean WL, Chaires JB, Olsen KW. "Inside-Out" PEGylation of Bovine β-Cross-Linked Hemoglobin. Artif Organs 2017; 41:351-358. [PMID: 28321886 DOI: 10.1111/aor.12928] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 01/12/2017] [Accepted: 01/13/2017] [Indexed: 01/06/2023]
Abstract
The development of a blood substitute is urgent due to blood shortages and potential communicable diseases. A novel method, inside-out PEGylation, has been used here to conjugate a multiarm maleimide-PEG (Mal-PEG) to β-cross-linked (βXL-Hb) hemoglobin (Hb) tetramers through the Cys β93 residues. This method produces a polymer with a single PEG backbone that is surrounded by multiple proteins, rather than coating a single protein with multiple PEG chains. Electrophoresis under denaturing conditions showed a large molecular weight species. Gel filtration chromatography and analytical ultracentrifugation determined the most prevalent species had three βXL-Hb to one Mal-PEG. Thermal denaturation studies showed that the cross-linked and PEGylated species were more stable than native Hb. Cross-linking under oxy-conditions produced a high oxygen affinity Hb species (P50 = 9.18 Torr), but the oxygen affinity was not significantly altered by PEGylation (P50 = 9.67 Torr). Inside-out PEGylation can be used to produce a hemoglobin-based oxygen carrier and potentially for other multiprotein complexes.
Collapse
Affiliation(s)
- Kyle D Webster
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL
| | - Dana Dahhan
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL
| | - Abigail M Otto
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL
| | - Cheyanne L Frosti
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL
| | | | - Jonathan B Chaires
- James Graham Brown Cancer Center.,Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Kenneth W Olsen
- Department of Chemistry and Biochemistry, Loyola University Chicago, Chicago, IL
| |
Collapse
|
24
|
Comparison of the Pharmacokinetic Properties of Hemoglobin-Based Oxygen Carriers. J Funct Biomater 2017; 8:jfb8010011. [PMID: 28335469 PMCID: PMC5371884 DOI: 10.3390/jfb8010011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/15/2017] [Accepted: 03/15/2017] [Indexed: 12/23/2022] Open
Abstract
Hemoglobin (Hb) is an ideal material for use in the development of an oxygen carrier in view of its innate biological properties. However, the vascular retention of free Hb is too short to permit a full therapeutic effect because Hb is rapidly cleared from the kidney via glomerular filtration or from the liver via the haptogloblin-CD 163 pathway when free Hb is administered in the blood circulation. Attempts have been made to develop alternate acellular and cellular types of Hb based oxygen carriers (HBOCs), in which Hb is processed via various routes in order to regulate its pharmacokinetic properties. These HBOCs have been demonstrated to have superior pharmacokinetic properties including a longer half-life than the Hb molecule in preclinical and clinical trials. The present review summarizes and compares the pharmacokinetic properties of acellular and cellular type HBOCs that have been developed through different approaches, such as polymerization, PEGylation, cross-linking, and encapsulation.
Collapse
|
25
|
Sickle Cell Disease, Severe Acidosis, and Inhalation Injury: Case Report of Neurologically Intact Survival After Cardiac Arrest From Smoke Inhalation. J Burn Care Res 2016; 38:e686-e688. [PMID: 27984409 DOI: 10.1097/bcr.0000000000000487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Smoke inhalation and carbon monoxide poisoning are a significant cause of mortality and neurologic morbidity. We present the unusual case of complete neurologic survival after prolonged hypoxia, severe acidosis, out of hospital cardiac arrest, and exposure to high levels of carbon monoxide poisoning in a patient with sickle cell disease. The hypothesis that there might be a potential protective effect from the type of hemoglobin seen in patients with sickle cell disease in carbon monoxide poisoning is discussed.
Collapse
|
26
|
Misra H, Bainbridge J, Berryman J, Abuchowski A, Galvez KM, Uribe LF, Hernandez AL, Sosa NR. A Phase Ib open label, randomized, safety study of SANGUINATE™ in patients with sickle cell anemia. Rev Bras Hematol Hemoter 2016; 39:20-27. [PMID: 28270341 PMCID: PMC5339363 DOI: 10.1016/j.bjhh.2016.08.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/03/2016] [Accepted: 08/22/2016] [Indexed: 02/05/2023] Open
Abstract
Background Treatment of sickle cell anemia is a challenging task and despite the well understood genetic and biochemical pathway of sickle hemoglobin, current therapy continues to be limited to the symptomatic treatment of pain, supplemental oxygen, antibiotics, red blood cell transfusions and hydroxyurea. SANGUINATE is a carbon monoxide releasing molecule and oxygen transfer agent under clinical development for the treatment of sickle cell anemia and comorbidities. Methods An open-label randomized Phase Ib study was performed in adult sickle cell anemia patients. Two dose levels of SANGUINATE were compared to hydroxyurea in 24 homozygotes for Hb SS. Twelve subjects received either a low dose (160 mg/kg) of SANGUINATE or 15 mg/kg hydroxyurea. Another 12 subjects received either a high dose (320 mg/kg) of SANGUINATE or 15 mg/kg hydroxyurea. The primary endpoint was the safety of SANGUINATE versus hydroxyurea in sickle cell anemia patients. Secondary endpoints included determination of the plasma pharmacokinetics and assessment of hematologic measurements. Results Musculoskeletal related adverse events were the most common. Transient troponin I levels increased in three patients, one of whom had an increase in tricuspid regurgitant velocity; however, no clinical signs were noted. Following an assessment of vital signs, tricuspid regurgitant velocity, electrocardiogram, serum biochemistry, hematology, urinalysis, and analysis of reported adverse events, SANGUINATE was found to be safe in stable sickle cell anemia patients. Conclusions The clinical trial met its primary objective of demonstrating an acceptable safety profile for SANGUINATE in patients with sickle cell anemia. This trial established the safety of SANGUINATE at both dose levels and permitted its advance to Phase II trials.
Collapse
Affiliation(s)
- Hemant Misra
- Prolong Pharmaceuticals, South Plainfield, United States.
| | | | - John Berryman
- Prolong Pharmaceuticals, South Plainfield, United States
| | | | | | | | | | | |
Collapse
|
27
|
Mullah SH, Abutarboush R, Moon-Massat PF, Saha BK, Haque A, Walker PB, Auker CR, Arnaud FG, McCarron RM, Scultetus AH. Sanguinate's effect on pial arterioles in healthy rats and cerebral oxygen tension after controlled cortical impact. Microvasc Res 2016; 107:83-90. [PMID: 27287870 DOI: 10.1016/j.mvr.2016.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/23/2016] [Accepted: 06/06/2016] [Indexed: 01/18/2023]
Abstract
Sanguinate, a polyethylene glycol-conjugated carboxyhemoglobin, was investigated for cerebral vasoactivity in healthy male Sprague-Dawley rats (Study 1) and for its ability to increase brain tissue oxygen pressure (PbtO2) after controlled cortical impact (CCI) - traumatic brain injury (TBI) (Study 2). In both studies ketamine-acepromazine anesthetized rats were ventilated with 40% O2. In Study 1, a cranial window was used to measure the diameters of medium - (50-100μm) and small-sized (<50μm) pial arterioles before and after four serial infusions of Sanguinate (8mL/kg/h, cumulative 16mL/kg IV), volume-matched Hextend, or normal saline. In Study 2, PbtO2 was measured using a phosphorescence quenching method before TBI, 15min after TBI (T15) and then every 10min thereafter for 155min. At T15, rats received either 8mL/kg IV Sanguinate (40mL/kg/h) or no treatment (saline, 4mL/kg/h). Results showed: 1) in healthy rats, percentage changes in pial arteriole diameter were the same among the groups, 2) in TBI rats, PbtO2 decreased from 36.5±3.9mmHg to 19.8±3.0mmHg at T15 in both groups after TBI and did not recover in either group for the rest of the study, and 3) MAP increased 16±4mmHg and 36±5mmHg after Sanguinate in healthy and TBI rats, respectively, while MAP was unchanged in control groups. In conclusion, Sanguinate did not cause vasoconstriction in the cerebral pial arterioles of healthy rats but it also did not acutely increase PbtO2 when administered after TBI. Sanguinate was associated with an increase in MAP in both studies.
Collapse
Affiliation(s)
- Saad H Mullah
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA.
| | - Rania Abutarboush
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA.
| | - Paula F Moon-Massat
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA.
| | - Biswajit K Saha
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA.
| | - Ashraful Haque
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA.
| | - Peter B Walker
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA.
| | - Charles R Auker
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA.
| | - Francoise G Arnaud
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA; Uniformed Services University of the Health Sciences, Department of Surgery, Bethesda, MD 20814, USA.
| | - Richard M McCarron
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA; Uniformed Services University of the Health Sciences, Department of Surgery, Bethesda, MD 20814, USA.
| | - Anke H Scultetus
- Naval Medical Research Center, NeuroTrauma Department, 503 Robert Grant Avenue Silver Spring, MD 20910, USA; Uniformed Services University of the Health Sciences, Department of Surgery, Bethesda, MD 20814, USA.
| |
Collapse
|
28
|
van den Born JC, Hammes HP, Greffrath W, van Goor H, Hillebrands JL. Gasotransmitters in Vascular Complications of Diabetes. Diabetes 2016; 65:331-45. [PMID: 26798119 DOI: 10.2337/db15-1003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In the past decades three gaseous signaling molecules-so-called gasotransmitters-have been identified: nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S). These gasotransmitters are endogenously produced by different enzymes in various cell types and play an important role in physiology and disease. Despite their specific functions, all gasotransmitters share the capacity to reduce oxidative stress, induce angiogenesis, and promote vasorelaxation. In patients with diabetes, a lower bioavailability of the different gasotransmitters is observed when compared with healthy individuals. As yet, it is unknown whether this reduction precedes or results from diabetes. The increased risk for vascular disease in patients with diabetes, in combination with the extensive clinical, financial, and societal burden, calls for action to either prevent or improve the treatment of vascular complications. In this Perspective, we present a concise overview of the current data on the bioavailability of gasotransmitters in diabetes and their potential role in the development and progression of diabetes-associated microvascular (retinopathy, neuropathy, and nephropathy) and macrovascular (cerebrovascular, coronary artery, and peripheral arterial diseases) complications. Gasotransmitters appear to have both inhibitory and stimulatory effects in the course of vascular disease development. This Perspective concludes with a discussion on gasotransmitter-based interventions as a therapeutic option.
Collapse
Affiliation(s)
- Joost C van den Born
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wolfgang Greffrath
- Department of Neurophysiology, Centre for Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | |
Collapse
|
29
|
Abuchowski A. PEGylated Bovine Carboxyhemoglobin (SANGUINATE™): Results of Clinical Safety Testing and Use in Patients. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 876:461-467. [DOI: 10.1007/978-1-4939-3023-4_58] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
30
|
Taguchi K, Yamasaki K, Seo H, Otagiri M. Potential Use of Biological Proteins for Liver Failure Therapy. Pharmaceutics 2015; 7:255-74. [PMID: 26404356 PMCID: PMC4588199 DOI: 10.3390/pharmaceutics7030255] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/17/2015] [Accepted: 08/26/2015] [Indexed: 01/11/2023] Open
Abstract
Biological proteins have unlimited potential for use as pharmaceutical products due to their various biological activities, which include non-toxicity, biocompatibility, and biodegradability. Recent scientific advances allow for the development of novel innovative protein-based products that draw on the quality of their innate biological activities. Some of them hold promising potential for novel therapeutic agents/devices for addressing hepatic diseases such as hepatitis, fibrosis, and hepatocarcinomas. This review attempts to provide an overview of the development of protein-based products that take advantage of their biological activity for medication, and discusses possibilities for the therapeutic potential of protein-based products produced through different approaches to specifically target the liver (or hepatic cells: hepatocytes, hepatic stellate cells, liver sinusoidal endothelial cells, and Kupffer cells) in the treatment of hepatic diseases.
Collapse
Affiliation(s)
- Kazuaki Taguchi
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan.
| | - Keishi Yamasaki
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan.
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan.
| | - Hakaru Seo
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan.
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan.
| | - Masaki Otagiri
- Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan.
- DDS Research Institute, Sojo University, 4-22-1 Ikeda, Nishi-ku, Kumamoto 862-0082, Japan.
| |
Collapse
|
31
|
Yin X, Wang X, Fan Z, Peng C, Ren Z, Huang L, Liu Z, Zhao K. Hyperbaric Oxygen Preconditioning Attenuates Myocardium Ischemia-Reperfusion Injury Through Upregulation of Heme Oxygenase 1 Expression: PI3K/Akt/Nrf2 Pathway Involved. J Cardiovasc Pharmacol Ther 2015; 20:428-38. [PMID: 25604781 DOI: 10.1177/1074248414568196] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 12/10/2014] [Indexed: 01/30/2023]
Abstract
BACKGROUND With the rise of the burden of ischemic heart disease, both clinical and economic evidence show a desperate need to protect the heart against myocardium ischemia-reperfusion injury-related complications following cardiac surgery or percutaneous coronary intervention. However, there is no effective intervention for myocardium ischemia-reperfusion injury as yet. METHODS We pretreated mice with 4 daily 2.0 absolute atmosphere (ATA) hyperbaric oxygen, then observed its effects on heart function parameters and infarct size following in situ ischemia-reperfusion. Multiple oxidative and inflammation products were measured in the myocardium. Next, we investigated the expression of heme oxygenase 1 (HO-1), phosphatidylinositol 3-kinase (PI3K)/serine/threonine protein kinase (Akt) pathway, and NF-E2-related factor 2 (Nrf2) in the presence of myocardium ischemia-reperfusion injury, hyperbaric oxygen preconditioning, and their inhibitors and their effects on heart function parameters. RESULTS Hyperbaric oxygen preconditioning ameliorated the cardiac function and histological alterations induced by myocardium ischemia-reperfusion injury, decreased oxidative products and proinflammatory cytokine. Hyperbaric oxygen preconditioning increased expression of HO-1, which was suppressed by PI3K inhibitor LY294002, Nrf2 knockout, and Akt inhibitor triciribine. The expression of Nrf2 was enhanced by hyperbaric oxygen preconditioning, but decreased by LY294002 and triciribine. The Akt was also activated by hyperbaric oxygen preconditioning but suppressed by LY294002. The hemodynamic assays showed that cardiac function was suppressed by LY294002, Nrf2 knockout, and triciribine. CONCLUSION These data present a novel signaling mechanism by which hyperbaric oxygen preconditioning protects myocardium ischemia-reperfusion injury via PI3K/Akt/Nrf2-dependent antioxidant defensive system.
Collapse
Affiliation(s)
- Xuesong Yin
- Department of Emergency Medicine, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Xiaofeng Wang
- Department of General Medicine, The Fourth Hospital of Heilongjiang Province, Harbin, People's Republic of China
| | - Zhixin Fan
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Chenghai Peng
- Department of Emergency Medicine, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Zhongqiao Ren
- Department of Emergency Medicine, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Le Huang
- Department of Emergency Medicine, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Zhuang Liu
- Department of Emergency Medicine, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| | - Kan Zhao
- Department of Emergency Medicine, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
32
|
Meng F, Tsai AG, Intaglietta M, Acharya SA. PEGylation of αα-Hb using succinimidyl propionic acid PEG 5K: Conjugation chemistry and PEG shell structure dictate respectively the oxygen affinity and resuscitation fluid like properties of PEG αα-Hbs. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2014; 43:270-81. [PMID: 24597567 DOI: 10.3109/21691401.2014.885443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PEGylation of intramolecularly crosslinked Hb has been studied here to overcome the limitation of dissociation of Hb tetramers. New hexa and deca PEGylated low oxygen affinity PEG-ααHbs have been generated. Influence of PEG conjugation chemistry and the PEG shell structure on the functional properties as well as PEGylation induced plasma expander like properties of the protein has been delineated. The results have established that in the design of PEG-Hbs as oxygen therapeutics, the influence of conjugation chemistry and the PEG shell structure on the oxygen affinity of Hb needs to be optimized independently besides optimizing the PEG shell structure for inducing resuscitation fluid like properties.
Collapse
Affiliation(s)
- Fantao Meng
- Hematology Division, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University , Bronx, NY , USA
| | | | | | | |
Collapse
|
33
|
Nielsen VG, Pretorius E. Carbon monoxide: Anticoagulant or procoagulant? Thromb Res 2013; 133:315-21. [PMID: 24360115 DOI: 10.1016/j.thromres.2013.12.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 11/25/2013] [Accepted: 12/02/2013] [Indexed: 11/18/2022]
Abstract
Within the past decade there have been several investigations attempting to define the impact of exogenous and endogenous carbon monoxide exposure on hemostasis. Critically, two bodies of literature have emerged, with carbon monoxide mediated platelet inhibition cited as a cause of in vitro human and in vitro/in vivo rodent anticoagulation. In contrast, interaction with heme groups associated with fibrinogen, α₂-antiplasmin and plasmin by carbon monoxide has resulted in enhanced coagulation and decreased fibrinolysis in vitro in human and other species, and in vivo in rabbits. Of interest, the ultrastructure of platelet rich plasma thrombi demonstrates an abnormal increase in fine fiber formation and matting that are obtained from humans exposed to carbon monoxide. Further, thrombi obtained from humans and rabbits have very similar ultrastructures, whereas mice and rats have more fine fibers and matting present. In sum, there may be species specific differences with regard to hemostatic response to carbon monoxide. Carbon monoxide may be a Janus-faced molecule, with potential to attenuate or exacerbate thrombophilic disease.
Collapse
Affiliation(s)
- Vance G Nielsen
- The Department of Anesthesiology, The University of Arizona College of Medicine, Tucson, AZ, USA.
| | - Etheresia Pretorius
- The Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| |
Collapse
|