Soluble Complement Receptor 1 Therapeutics

Matthew P. Hardy1*, Tony Rowe1, and Sandra Wymann2

1CSL, Bio21 Institute, Victoria, Australia

2CSL, CSL Biological Research Centre, Bern, Switzerland


Human Complement Receptor 1 (CR1/CD35) is a potent negative regulator of the complement system. Its mechanism of action is through interaction with the complement activation fragments, C3b and C4b to mediate decay acceleration of the C3 and C5 convertase complexes as well as cleavage of both ligands into inactive fragments via cofactor activity. The result is inhibition of the classical, lectin, and alternative complement pathways. This article will focus on recombinant soluble forms of CR1 that have been generated as potential therapeutics for complement-mediated disorders. Specifically, we will review and contrast the in vitro and in vivo properties of: sCR1 (BRL55730/TP10/CDX-1135), the soluble full-length extracellular domain of human CR1; sCR1-sLex (TP20), a glyco-engineered version of sCR1 additionally targeted to activated endothelium; APT070 (Mirococept), a CR1 fragment conjugated to a myristoylated peptide to enhance tissue targeting; and CSL040, a soluble truncated version of the CR1 extracellular domain which exhibits altered potency and pharmacokinetic properties as compared to the parental molecule. The data obtained from studies on the effects of these CR1-based molecules in animal models of disease and their therapeutic applications will also be discussed.


Introduction

The complement cascade is an arm of the innate immune system that has evolved as a primary defence mechanism against pathogens and other deleterious processes. It consists of multiple proteins found both in plasma and at the surface of many cell types which work to target not only immune complexes and cells for phagocytosis via the process of opsonization, but also to drive inflammation via the formation of the anaphylatoxins C3a and C5a, and to initiate cell lysis directly through the generation of the membrane attack complex. These processes are initiated by the classical, lectin, and alternative complement pathways by a variety of activating factors such as immune complexes, endotoxin, neoepitopes, specific carbohydrate moieties, and by the spontaneous C3 ‘tick-over’ mechanism1-4. To prevent unchecked activation of the complement cascade and damage to host cells or tissues, specific proteins have evolved as regulators of complement. Both transmembrane and soluble forms of these regulators exist, acting at all levels of the complement cascade. Examples include CD465, CD556, CD597, Factor H8, Factor I9, and the complement receptors CR1 – CR410.

Complement Receptor 1

Human CR1 (CD35) is a central regulator of the complement system, acting at the level of complement component C3 to inhibit the classical, lectin and alternative pathways11. It is primarily a cell surface bound membrane protein expressed on erythrocytes (E-CR1) and immune cell types such as monocytes, macrophages, neutrophils, eosinophils, certain B and T cells, and glomerular podocytes11-13. A soluble version of CR1 exists; this is produced at very low levels (approximately 50 ng/mL) by proteolytic cleavage of the extracellular domain from the membrane, rather than produced constitutively14,15. CR1 belongs to a family of complement receptors that include CR2, CR3 and CR410. It also belongs to a wider family of structurally related proteins referred to as the Regulators of Complement Activation. These include CD46, CD55, C4-binding protein, and Factor H, and are characterized by repetitive and highly homologous modules of approximately 60 amino acids known as short consensus repeat (SCR) domains16. The dominant allelic variant of human CR1 (Figure 1) is a 2039 amino acid protein with a 41 amino acid signal peptide, a 1930 amino acid extracellular domain, and a short cytoplasmic tail16,17. The extracellular domain of human CR1 is further comprised of 30 SCR domains which are themselves arranged into four long homologous repeat domains (LHR-A, -B, -C, -D), each composed of seven SCR domains16,18,19. Although SCR domains 29-30 are not technically part of LHR-D, they are often included for experimental purposes20-22. There is a high degree of homology (up to 99%) between SCR domains within CR116,23, such as the SCR8-10 and SCR15-17 domains located within LHR-B and LHR-C, respectively (Figure 1). Several allelic variants of human CR1 exist that contain fewer or additional LHR domains16,17. CR1 has a monomeric and highly flexible structure similar to other family members and adopts a “string of pearls” type conformation as suggested by its domain structure, negative stain microscopy, X-ray scattering and analytical ultracentrifugation24-26. CR1 is also highly glycosylated on multiple glycan sites found within its primary amino acid sequence, with its glycan structure determined to be exclusively N-linked27.

JISS-22-1240-fig1

Figure 1. The structure of human CR1: Shown here schematically, human CR1 is a type I membrane protein composed of an N-terminal extracellular domain, a transmembrane region (TM), and a short cytoplasmic tail. CR1 also contains a signal peptide (SP) that is removed upon expression of the mature protein on the cell surface or when soluble CR1 is expressed recombinantly. The extracellular domain is made up of four long homologous repeat (LHR) domains, labelled –A to –D. The numbering above the top schema refers to the number of the last amino acid of each LHR domain and the final amino acid of the cytoplasmic region (based on Met+1), with the exception of “42”, which refers to the number of the first amino acid of LHR-A. The lower schema shows how each LHR domain is comprised of a number of short consensus repeat (SCR) domains to a total of 30 within the extracellular domain of CR1. The green and red bracketed regions with the numerated percentages denote the degree of amino acid homology between sets of SCR domains located within LHR-A and LHR-B. At the bottom of the figure the location (horizontal bars) and details of the ligand binding, decay acceleration, and co-factor activity properties of CR1 are shown.

The biological activity of human CR1 is mediated by binding to the ligands C3b and C4b, which are activated fragments of C3 and C4 zymogens, respectively12,28. Since they share similar binding sites on C3b and C4b, CR1 competitively displaces the Factor Bb and C2a catalytic fragments from the C3 convertases (C4bC2a, C3bBb) and C5 convertases (C4bC2aC3b, C3bBbC3b) which are formed upon complement activation. This mechanism of action is termed decay acceleration activity (DAA) and works to block further complement activation at the cell surface29,30. Human CR1 has a second function which is to act as a co-factor for complement Factor I, a serum protease that can cleave CR1-bound C3b and C4b via this co-factor activity (CFA) into the inactive forms iC3b and iC4b/C4c, to further inhibit complement activation13,29-32. Weak binding of human CR1 to iC3b and cleavage to further degradation products (C3c and C3dg) mediated by Factor I has also been reported24,31,33.

Soluble Complement Receptor 1 Therapeutics

The structure of human CR1 as a type I membrane protein makes it particularly amenable to engineering to create soluble versions via removal of the transmembrane and cytoplasmic regions, plus any part of the extracellular domain deemed undesirable. Soluble CR1 proteins have been used for more than 30 years as negative regulators of the complement system in preclinical and clinical studies of the effects of complement inhibition in various disease and injury settings. Here we review four of them in detail: sCR1 (also known as BRL55730, TP10, and CDX-1135), sCR1-sLex (TP20), APT070 (Mirococept), and finally a recently identified and characterized molecule, CSL040 (Figure 2).

JISS-22-1240-fig2

Figure 2. Therapeutic agents based on human CR1: Shown here schematically are the structures of four soluble CR1-based recombinant therapeutics: A) sCR1, also known as BRL55730, TP10, or CDX-1135; B) sCR1-sLex, also called TP20; C) APT070 (Mirococept); D) CSL040. Each LHR domain found within each molecule is indicated with the exception of APT070 which has its SCR domains denoted. The vertical lines and numbering above the schemas shown in A, B and D denote the position and amino acid numbering (based on Met+1) of N-linked glycosylation sites. The red vertical lines capped by diamonds in panel B represent the sialyl Lewis X glycans found on TP20. The red “C” at the C-terminal end of SCR3 in panel C denotes the presence of a C-terminal cysteine on APT070 to which is conjugated the myristoylated peptide (wavy line) needed for tissue targeting.

sCR1 (BRL55730 / TP10 / CDX-1135)

The first description of a soluble recombinant molecule designed as a potential therapeutic inhibitor of complement was published in 199024,34. Using recently developed molecular biology techniques, it was possible to synthesize the cDNA encoding the entire extracellular domain of CR1 containing all four LHR domains (Figure 2A) and to express this protein recombinantly. Studies then showed that sCR1 retained all the biological functions of its parental molecule, such as ligand binding to C3b and C4b, Factor I-mediated CFA, C3 and C5 convertase DAA, and potent inhibition of all three complement pathways in vitro20,22,24,34-36. Broad cross-reactivity across multiple surrogate species also meant that sCR1 was particularly suitable for studies in animals to assess its pharmacokinetic and pharmacodynamic properties, as well as assessing its potency in vivo in animal models of disease or injury where it was hypothesized that complement played a role in the aetiology and/or progression of cellular or tissue damage. However, the in vivo half-life of sCR1 in rats was determined to be approximately 100 minutes37,38. This relatively short half-life limited its use to acute, rather than chronic settings.

This short half-life of sCR1 in vivo has not prevented its widespread use in a host of animal models for proof-of-concept testing, and we were able to identify over one hundred different studies in which it has been tested (Table 1). A breakdown of these studies is shown in Figure 3 where sCR1 has been applied to animal models across multiple therapeutic areas such as inflammation, tissue injury, neurology, auto-immunity and immune-complex mediated diseases, and infection, with a particular emphasis on ischemia-reperfusion injury (IRI) and transplantation (Figure 3A; Table 1). Several different animal species have been employed in these in vivo proof-of-concept studies with sCR1 (Figure 3B; Table 1), with most studies performed in rats as a surrogate species where the strength of the complement system is similar to that of humans39. Other important information that has been gained from these animal studies has revolved around dosing and tolerability, which would have informed subsequent human studies. Soluble CR1 has been found to be safe and well tolerated across species at doses – both single and multiple – of up to 60 mg/kg, and various routes of administration such as intravenous, intraperitoneal, and even intrathecal have been safely employed (Table 1). Most importantly, sCR1 has been found to be protective in the vast majority of studies in which it has been tested (Figure 3C; Table 1), although it has been applied mostly prophylactically, rather than therapeutically, potentially limiting the translatability of data to human disease settings. In vivo studies weren’t the only means to assess the suitability of sCR1 for clinical applications. Two early studies showed that sCR1 is a potent inhibitor of membrane-induced complement activation ex vivo40,41, rendering it of potential clinical utility during haemodialysis.

JISS-22-1240-fig3

Figure 3. The use of sCR1 (BRL55730 / TP10 / CDX-1135) in animal models:Graphical representation in pie chart form of some of the key characteristics of the 101 animal models used to test sCR1 in vivo and the proportions of their usage. A) The indications in which sCR1 was tested, separated broadly into therapeutic areas (see key below chart). IRI: ischemia-reperfusion injury; IC: immune complex; CPB: Cardiopulmonary bypass. B) The species in which sCR1 was tested. When Xenografts required the use of two species, both were counted. C) The use of sCR1 prophylactically or therapeutically, and whether that usage was protective (as defined by statistically significant differences in key disease biomarkers compared to controls) or had no significant effect. The degree of protection is not noted here, as this varies and is defined differently from model to model. See Supplementary table I for the complete list and details of each animal model used to assess in vivo efficacy of sCR1.

Table 1: Use of sCR1 (BRL55730 / TP10 / CDX-1135) in vivo and ex vivo – animal models

Species

Experiment / model

Route(s) of administration

Dose(s)

Prophylactic or Therapeutic

Effecta

Reference

Ischemia-Reperfusion Injury (IRI)

Rat

Myocardial IRI

intravenous

1 mg/rat

Prophylactic

Protective

Weisman et al (1990a)34

Weisman et al (1990b)24

Rat

Intestinal IRI

intravenous

3, 6 mg/rat x4

Prophylactic

Protective

Hill et al (1992)42

Rat

Hind Limb IRI

intravenous

1, 3, 6 mg/rat

Prophylactic

Protective

Lindsay et al (1992)43

Mouse

Skeletal (Cremaster) Muscle IRI

intravenous

100 ug/mouse

+ 100 ug/hr/mouse infusion

Prophylactic

Protective

Pemberton et al (1993)44

Ratb

Cardiac IRI

Perfusion

10 μg/mL

Prophylactic

Protective

Shandelya et al (1993)45

Rat

Myocardial IRI

intravenous

5 mg/kg

Prophylactic

Protective

Smith et al (1993)46

Rat

Liver IRI

intravenous

25 mg/kg x1, or 50 mg/kg x2

Prophylactic

Protective

Chavez-Cartaya et al (1995)47

Rat

Intestinal IRI

intravenous /

Perfusion

20 mg/kg /

0.286 mg/mL

Prophylactic

Protective

Xiao et al (1997)48

Mouse

Intestinal IRI

intravenous

10 mg/kg

Prophylactic

Protective

Austen et al (1999)49

Rat

Intestinal IRI

intravenous

12 mg/kg x 2

Prophylactic

Protective

Eror et al (1999)50

Rat

Intestinal IRI

intravenous

12 mg/kg

Therapeutic

Protective

Eror et al (1999)50

Mouse

Intestinal IRI

intravenous

20 mg/kg

Prophylactic

Protective

Williams et al (1999)51

Rat

Acute Myocardial Infarction

intravenous

1, 5, 15 mg/kg

Prophylactic

Protective

Zacharowski et al (1999)52

Rat

Hepatic IRI

intravenous

15 mg/kg

Prophylactic

Protective

Lehmann et al (2001)53

Rat

Pancreatic IRI

intravenous

15 mg/kg

Prophylactic

Protective

von Dobschuetz et al (2004)54

Rat

Placental Ischemia

intravenous

15 mg/kg/day

Prophylactic

Protective

Lillegard et al (2013)55

Regal et al (2019)56

Mouse

Renal IRI

intravenous

25 mg/kg

Prophylactic

Protective

Hameed et al (2020)57

Transplantation

Guinea Pig to Rat

Cardiac Xenograft

intravenous

3, 5.9, 15, 60 mg/kg

Prophylactic

Protective

Pruitt et al (1991)58

Guinea Pig to Rat

Renal Xenograft

intravenous

50 mg/kg

Prophylactic

Protective

Chrupcala et al (1994)59

Guinea Pig to Rat

Cardiac Xenograft

intravenous

20 mg/kg

Prophylactic

Protective

Zehr et al (1994)60

Pig to Cyno

Cardiac Xenograft

intravenous

15 mg/kg

Prophylactic

Protective

Pruitt et al (1994)61

Guinea Pig to Rat

Cardiac Xenograft

intravenous

25 mg/kg + 20mg/kg/12hr (repeat), or 25mg/kg + 40 mg/kg/day (infusion)

Prophylactic

Protective

Candinas et al (1996)62

Pig to Cyno

Cardiac Xenograft

intravenous

25 mg/kg + 40 mg/kg/day

Prophylactic

Protective

Davis et al (1996)63

Rat

Lung Allograft

intravenous

25 mg/kg/day

Prophylactic

Protective

Pratt et al (1996a)64

Pratt et al (1996b)37

Pratt et al (1997)65

Guinea Pig to Rat

Cardiac Xenograft

intravenous

20 mg/kg

Prophylactic

Protective

Fujiwara et al (1997)66

Rat

Lung Allograft

intravenous

15 mg/kg

Prophylactic

Protective

Naka et al (1997)67

Pig to Cyno

Cardiac Xenograft

intravenous

25 mg/kg + 40 mg/kg/day

Prophylactic

Protective

Pruitt et al (1997)68

Rat

Liver Allograft

intravenous

15 mg/kg

Prophylactic

Protective

Lehmann et al (1998)69

Pig

Lung Allograft

intravenous

15 mg/kg

Prophylactic

Protective

Pierre et al (1998)70

Pig

Lung Allograft

intravenous

15 mg/kg

Prophylactic

Protective

Schmid et al (1998)71

Pig to Cyno

Intraportal Xenograft

intravenous

40 mg/kg

Prophylactic

Protective

Bennet et al (2000)72

Lundgren et al (2001)73

Rat

Tracheal Allograft

intraperitoneal

20 mg/kg/day

Prophylactic

Protective

Kallio et al (2000)74

Rat

Lung allograft

intracardiac

10 mg/kg

Prophylactic

Protective

Stammberger et al (2000)75

Schmid et al (2001)76

Pig to Cyno

Renal Xenograft

intravenous

40 mg/kg + (17-20 mg/kg Daily)

Prophylactic

No effect

Lam et al (2005)77

Rat

Renal Allograft

intravenous

25 mg/kg

Prophylactic

Protective

Damman et al (2011)78

Other Injury

Rat

CVF-induced Lung Injury

intravenous

5, 10, 15, 20, 25 mg/kg

Prophylactic

Protective

Mulligan et al (1992)79

Rat

Thermal Injury

intravenous

5, 10, 15, 20, 25 mg/kg

Therapeutic

Protective

Mulligan et al (1992)79

Rat

LPS-induced Lung Injury

intravenous

1, 10 mg/kg

Prophylactic

Protective

Rabinovici et al (1992)80

Rabbitb

NHS-induced Cardiac Injury

Perfusion

20 nM

Prophylactic

Protective

Homeister et al (1993)81

Pigb

Human blood-induced Cardiac Injury

Perfusion

70, 300 μg/mL

Prophylactic

Protective

Pruitt et al (1994)61

Rat

IL2-induced Lung Injury

intravenous / intraperitoneal

10, 30 mg/kg (50% each route)

Prophylactic

Protective

Rabinovici et al (1994)82

Rabbitb

Human Plasma-induced Cardiac injury

Perfusion

20 mM

Prophylactic

Protective

Gralinski et al (1996)83

Rat

Acid-induced Lung Injury

intravenous

10 mg/kg

Prophylactic

Protective

Nishizawa et al (1996)84

Rat

Acid-induced Lung Injury

intravenous

10 mg/kg

Therapeutic

Protective

Nishizawa et al (1996)84

Mouse

Acid-induced Lung Injury

intravenous

20 mg/kg

Prophylactic

Protective

Weiser et al (1997)85

Guinea Pig

Porphyrin-induced Phototoxicity

intraperitoneal

60 + 20/40 mg/kg

Prophylactic

Protective

Nomura et al (1998)86

Rat

CVF-induced lung injury

intravenous

0.3, 1.5, 4.5 mg/rat

Prophylactic

Protective

Mulligan et al (1999)87

Rabbitb

NHS-induced Lung Injury

Perfusion

2.0 ug/mL

Prophylactic

Protective

Heller et al (2000)88

Rabbitb

NHS-induced Cardiac Injury

Perfusion

20 nM

Prophylactic

Protective

Tanhehco et al (2000)89

Mouse

Acid-induced Lung injury

intravenous

5, 10 mg/kg

Prophylactic

Protective

Kyriakides et al (2001)90

Mouse

Acid-induced Lung injury

intravenous

10 mg/kg

Therapeutic

Protective

Kyriakides et al (2001)90

Pigb

Human blood-induced Lung Injury

Perfusion

100 μg/mL

Prophylactic

No effect

Azimzadeh et al (2003)91

Pfeiffer et al (2005)92

Mouseb

Human blood-induced Lung Injury

Perfusion

225 μg/mL

Prophylactic

Protective

Schroder et al (2003)93

Ratc

Hypertension and Renal Injury

intraperitoneal

15 mg/kg/day

Prophylactic

No effect

Regal et al (2018)94

Ratc

Hypertension and Renal Injury

intraperitoneal

15 mg/kg/day

Therapeutic

No effect

Regal et al (2018)94

Immune complex & Inflammation

Rat

Reverse Passive Arthus Reaction

intradermal

0.003, 0.03, 0.3, 1, 3, 30 μg/site

Prophylactic

Protective

Yeh et al (1991)95

Rat

Glycogen-induced Peritonitis

intravenous

7.5 mg/kg x2

Therapeutic

Protective

Mulligan et al (1992)79

Rat

IgG immune complex Alveolitis

intravenous

3.75 mg/kg x4

Prophylactic

Protective

Mulligan et al (1992)79

Rat

TNF- and Collagen-induced Arthritis

intraperitoneal

20 mg/kg/day

Prophylactic

Protective

Fava et al (1993)96

Rat

Anti-Thy1 Glomerulonephritis

intraperitoneal

60 mg/kg/day

Prophylactic

Protective

Couser et al (1995)97

Rat

Anti-Concavalin-A Glomerulonephritis

intraperitoneal

60 mg/kg/day

Prophylactic

Protective

Couser et al (1995)97

Rat

Passive Heymann Nephritis

intraperitoneal

60 mg/kg/day

Prophylactic

Protective

Couser et al (1995)97

Rat

Experimental Autoimmune Thyroiditis

intraperitoneal

10 mg/kg/day

Prophylactic

No effect

Metcalfe et al (1996)98

Rat

Cerulein-Induced Pancreatitis

intravenous

15 mg/kg/hour

Prophylactic

No effect

Weiser et al (1996)99

Rat

Cerulein-Induced Pancreatitis

intravenous

22.5 mg/kg

Prophylactic

Protective

Acioli et al (1997)100

Rat

Mono-articular Arthritis

intravenous and/or

intra-articular

20 mg/kg/day

 and/or 200 μg/joint

Prophylactic

Protective

Goodfellow et al (1997)101

Rat

Mono-articular Arthritis

intra-articular

200 μg/joint

Therapeutic

No effect

Goodfellow et al (1997)101

Rat

Antigen-induced Pleural Inflammation

intravenous

10 mg/kg or 15 mg/kg x2

Prophylactic

Protective

Lima et al (1997)102

Rat

Muscle Inflammation

intraperitoneal

20 mg/kg + (10 mg/kg x 3)

Prophylactic

Protective

Frenette et al (2000)103

Rat

Collagen-induced Arthritis

intravenous

15 mg/kg twice daily

Prophylactic

Protective

Goodfellow et al (2000)104

Rat

Collagen-induced Arthritis

intravenous

15 mg/kg twice daily

Therapeutic

Protective

Goodfellow et al (2000)104

Rat

Acute Arthritis/Synovitis

intra-articular

0.5 mg/joint

Prophylactic

Protective

Mizuno et al (2000)105

Rat

Acute Arthritis/Synovitis

intravenous

20 mg/kg

Prophylactic

No effect

Mizuno et al (2000)105

Rat

Allergic Asthma

intraperitoneal

10 mg/kg

Prophylactic

Protective

Abe et al (2001)106

Rat

Thrombotic Glomerulonephritis

intravenous

20 mg/kg

Prophylactic

Protective

Kondo et al (2001)107

Rat

Severe Acute Pancreatitis

intravenous

12 mg/kg x 2

Prophylactic

Protective

Hartwig et al (2006)108

Moused

C3 Glomerulonephritis

intraperitoneal

25 or 50 mg/kg/day

Prophylactic

Protective

Zhang et al (2013)109

Cardiopulmonary Bypass

Pig

Cardiopulmonary Bypass

intravenous

6 mg/kg x2

Prophylactic

Protective

Gillinov et al (1993)110

Pig

Cardiopulmonary Bypass

intravenous

10 mg/kg

Prophylactic

Protective

Lazar et al (1999)111

Pig

Cardiopulmonary Bypass

intravenous

10 mg/kg

Prophylactic

Protective

Chai et al (2000)112

Neurology

Rat

Antibody-mediated demyelinating

Experimental Allergic Encephalomyelitis

intraperitoneal

20 mg/kg/day

Prophylactic

Protective

Piddlesden et al (1994)113

Rat

Experimental Autoimmune Neuritis

intraperitoneal

30 mg/kg/day

Therapeutic

Protective

Jung et al (1995)114

Rat

Traumatic Brain Injury

intravenous / intraperitoneal

20 mg/kg + 15 mg/kg

Prophylactic

Protective

Kaczorowski et al (1995)115

Rat

Experimental Autoimmune Myasthenia Gravis

intraperitoneal

20 mg/kg/day

Prophylactic

Protective

Piddlesden et al (1996)116

Rat

Experimental Allergic Neuritis

intraperitoneal

60 mg/kg x 2

Therapeutic

No effect

Vriesendorp et al (1997)117

Mouse

Stroke (middle cerebral artery)

intravenous

15 mg/kg

Therapeutic

Protective

Huang et al (1999)118

Rat

Sciatic Inflammatory Neuropathy

intrathecal

50 μg/rat

Therapeutic

Protective

Twining et al (2005)119

Rat

Chronic Constriction Nerve Injury

intrathecal

50 μg/rat

Therapeutic

Protective

Twining et al (2005)119

Rat

gp120-induced Mechanical Allodynia

intrathecal

50 μg/rat

Therapeutic

Protective

Twining et al (2005)119

Baboon

Reperfused Stroke

intravenous

15 mg/kg

Prophylactic

No effect

Mocco et al (2006)120

Rat

Nerve Crush Injury

intraperitoneal

15 mg/kg/day

Prophylactic

Protective

Ramaglia et al (2008)121

Ramaglia et al (2009)122

Rat

Spinal Cord Injury

intravenous

6 mg/kg/day

Therapeutic

Protective

Li et al (2010)123

Infection & Shock

Guinea Pig

Anaphylaxis

(Passive and active)

intravenous / intraperitoneal

15, 105 mg/kge

Prophylactic

Protective

Regal et al (1993)124

Rat

Bacterial infection

intravenous

10, 25 mg/kg

Prophylactic

Protective

Swift et al (1994)125

Rat

Haemorrhage / Resuscitation

intravenous

15 mg/kg

Prophylactic

Protective

Fruchterman et al (1998)126

Rat

Haemorrhage / Resuscitation

intravenous

15 mg/kg

Prophylactic

Protective

Spain et al (1999)127

Guinea Pig

Forssman (pulmonary) Shock

intravenous

15 mg/kg

Prophylactic

Protective

Wagner et al (1999)128

Rat

Acute Shock

intravenous

20 mg/kg

Prophylactic

Protective

Mizuno et al (2002)129

Rat

Acute Shock

intravenous

20 mg/kg

Therapeutic

Protective

Mizuno et al (2002)129

Mouse

Red Blood Cell Transfusion

intravenous

1.5, 10 mg/kg

Prophylactic

Protective

Yazdanbakhsh et al (2003)130

Rat

Haemorrhagic Shock

intravenous

No dose provided

Prophylactic

Protective

Chen et al (2016)131

Listings are separated into broad therapeutic areas for ease of reading. Some animal models fit into more than one category; in these cases, the most appropriate was chosen. While every attempt has been made to capture all reported usage of sCR1 in vivo and ex vivo from extant literature, there may be examples that have been missed. aThe degree of protection varies from model to model but must be statistically significant compared to controls to be deemed protective. bExperiment performed ex vivo. cDahl Salt-sensitive (SS) rats. dComplement Factor H knock-out mice (a model of C3 Glomerulonephritis) with or without the transgene for human CR1. e15 mg/kg dose administered intravenously only; 105 mg/kg dose administered cumulatively both intravenously and intraperitoneally over a 24 hr dosing period, rather than as a single dose. IRI: Ischemia-reperfusion injury; CVF: Cobra Venom Factor; LPS: Lipopolysaccharide; TNF: Tumour Necrosis Factor; NHS: Normal Human Serum; Cyno: Cynomolgus monkey. The multipliers added to entries within the Dose(s) column refer to the number of doses used that were administered less frequently than daily.

The strength of the sCR1 in vivo data from animal models led to the initiation of human clinical trials as summarised in Table 2. A Phase I single ascending dose study132 of sCR1 (TP10) in acute lung injury and acute respiratory distress syndrome patients demonstrated its safety and tolerability, with a pharmacokinetic/pharmacodynamic assessment showing an in vivo half-life of approximately 70 hours and complete inhibition of complement ex vivo in hemolytic assays at doses >1 mg/kg. Similar results were observed in a subsequent Phase I/II study in infants undergoing cardiopulmonary bypass (CPB), with evidence suggesting that sCR1 could confer some clinical benefit in this setting133. Given these tentative findings and previous data from pig models of CPB110-112 , a Phase II clinical study with 564 patients undergoing CPB during cardiac surgery was then initiated to determine the efficacy of sCR1 (TP10) in this indication. Unfortunately, the outcome of this clinical trial reported no overall efficacy in this indication for sCR1134 despite showing complete inhibition of complement. When the data from the study was assessed in more detail, there appeared to be a gender-specific effect, with males showing significant improvements in the primary endpoints compared to both females and controls134. A follow-on study of sCR1 (TP10) in female patients undergoing CPB and administered sCR1 (TP10) showed no effect135. Of equal interest is a study in a single patient who inadvertently received an ABO-mismatched lung allograft, where compassionate use of sCR1 (TP10) administration suggested some efficacy136. This led to a Phase II clinical study conducted in 59 patients undergoing lung transplantation, in which sCR1 (TP10) showed significant positive effects on extubation times and time of stay in intensive care, although no significant differences in other parameters such as death and graft rejection rates were found137. More recent attempts to use sCR1 clinically have been made (Table 2), but these failed for various reasons, and it appears that the entire program has been discontinued138.

Table 2: Use of sCR1 (TP10 / CDX-1135) in vivo – human studies

Clinical Trial

Patient number / indication

Route of administration

Dose(s)

 Main Outcomes

Reference

Phase I

24

Acute Lung Injury and ARDS

intravenous

0.1, 0.3, 1.0, 3.0, 10 mg/kg

Single ascending dose safe and well tolerated

Half-life of 69 hours

Doses >1 mg/kg inhibit Complement activity

Zimmerman et al (2000)132

Case Report

1

ABO-mismatched Lung Allograft

intravenous

15 mg/kg x 5 doses

every 3-4 days

Reduction of anti-A antibody titer

No humoral injury or cellular rejection

Patient stable for 3 years post-transplant

Pierson et al (2002)136

Phase I/II

15 (infants <1 year old)

Cardiopulmonary Bypass

intravenous

10 mg/kg plus 0.1 mg/mL to bypass circuit

All infants survived and no TP10-related adverse events

Half-life of 71 hours

TP10 may protect against increase in vascular permeability

Li et al (2004)133

Phase II

564

Cardiopumonary Bypass

intravenous

1, 3, 5, or 10 mg/kg

Elimination half-life of 55-57 hours; complement activity inhibited

No effect on primary end point between TP10 and controls

Significant improvement in endpoints in males only

Lazar et al (2004)134

Phase II

297 (Females)

Cardiopulmonary Bypass

intravenous

10 mg/kg

TP10 well tolerated

Effective inhibition of complement

No effect on primary end points of death or myocardial infarction

Lazar et al (2007)135

Phase III

59

Lung Allograft

intravenous

10 mg/kg

Significant increase in patients undergoing early extubation

Total time on ventilator and in intensive care shorter

No difference in operative deaths, infection and rejection rates

Keshavjee et al (2005)137

Phase I

0

C3 Glomerulopathy

-

-

ClinicalTrials.gov Identifier: NCT02302755

Study withdrawn (2014) – no recruitment

-

Phase I

0

Dense Deposit Disease

-

-

ClinicalTrials.gov Identifier: NCT01791686

Study Terminated (2014) – due to slow enrolment, portfolio prioritization and issues with role of complement in indication

-

sCR1-sLex (TP20)

Glyco-engineering recombinant proteins to improve or modify their biological activity has been used for many years139. One such approach was used to generate a glyco-engineered variant of sCR1, denoted sCR1-sLex (TP20). Specific Sialyl-Lewis-X (sLex) tetra-saccharide carbohydrate motifs are found on the glycans of neutrophils and other leucocytes, and are made up of N-acetylglucosamine, galactose, neuraminic acid and fucose. These bind to specific ligands called selectins, particularly P-, L- and E-selectin, expressed on the surface of vascular endothelium140,141. Since leucocyte migration mediated by selectin binding contributes to inflammation and tissue damage, and given that selectins can be upregulated during disease142,143, it was thought that a bi-functional molecule (sCR1-sLex; Figure 2B) able to both inhibit complement and act as a selectin antagonist could be generated by decorating the N-Glycans of sCR1 with sLex motifs. There would be the added benefit of targeting sCR1 to the site of tissue damage144. Earlier studies have also demonstrated the ability of sLex to act as a stand-alone therapeutic, reducing neutrophil infiltration into tissue and protecting against damage145,146.

Engineering soluble CR1 to specifically expresses the sLex motif on its N-glycans was not easily achieved, requiring the use of a Chinese Hamster Ovary (CHO) cell line – LEC11 – to express the α(1,3)-fucosyltransferase needed to add α(1,3)-fucose to the N-glycans present on sCR1144,147,148. The end-result, sCR1-sLex, was shown to have a 10:1 ratio of sLex to sCR1 with an increased sialic acid content as well as a doubling of fucose content144. In vitro assessment of sCR1-sLex demonstrated increased binding to CHO cell-expressed E-selectin relative to its unmodified counterpart, and a dose-dependent blockade of U937 cell adhesion to immobilized P-selectin-IgG. Unmodified sCR1 was ineffective in the latter assay. It should be noted that sCR1-sLex also exhibited a small (<2-fold) but statistically significant decrease in complement inhibitory activity compared to sCR1 alone144.

Over the next few years, sCR1-sLex was tested in a number of animal disease models, several of which also employed sCR1 as a comparator, to determine its potency in vivo (See Table 3). The first model tested was a mouse model of middle cerebral artery occlusion (stroke) in which complement was shown to play a role118. In this model, sCR1 and sCR1-sLex were compared in both prophylactic and therapeutic settings, with equal doses of 15 mg/kg administered. In these experiments, sCR1-sLex was found to be more efficacious than sCR1, showing improvements in animal survival and additional reductions in infarct volume, neural deficit score and intracerebral hemorrhage. In an experimental rat model of cobra venom factor-induced lung injury, sCR1-sLex was almost twice as protective as sCR1, with dose-dependent reductions in both vascular leakage and lung neutrophil accumulation measured87. Of additional note in this study was the binding of sCR1-sLex to the lung vasculature, a phenomenon not observed with sCR1 alone. Addition of an anti-P-selectin antibody blocked this interaction, demonstrating the specificity of the binding of sCR1-sLex to P-selectin.

Table 3: Use of sCR1-sLex (TP20) in vivo – animal models

Species

Experiment / model

Route of administration

Dose(s)

Prophylactic or Therapeutic

Effecta

References

Mouse

Stroke (middle cerebral artery)

intravenous

15 mg/kg

Prophylactic

Protective

Huang et al (1999)118

Mouse

Stroke (middle cerebral artery)

intravenous

15 mg/kg

Therapeutic

Protective

Huang et al (1999)118

Rat

CVF-induced lung injury

intravenous

0.3, 1.5, 4.5 mg/rat

Prophylactic

Protective

Mulligan et al (1999)87

Rat

Acute Myocardial Infarction

intravenous

1, 5, 15 mg/kg

Prophylactic

Protective

Zacharowski et al (1999)52

Rat

Lung allograft

intra-cardiac

10 mg/kg

Prophylactic

Protective

Stammberger et al (2000)75

Schmid et al (2001)76

Mouse

Acid-induced Lung injury

intravenous

5, 10 mg/kg

Prophylactic

Protective

Kyriakides et al (2001)90

Mouse

Acid-induced Lung injury

intravenous

10 mg/kg

Therapeutic

Protective

Kyriakides et al (2001)90

Baboon

Reperfused stroke

intravenous

15 mg/kg

Prophylactic

Worse outcomeb

Ducruet et al (2007)149

Zacharowski et al52 tested sCR1-sLex in a rat model of myocardial IRI, showing reductions in both infarct size, cardiac troponin T release (a marker of cardiac tissue damage) and polymorphonuclear cell infiltrate at administered doses of 1 and 5 mg/kg, although a 15 mg/kg dose showed no further reduction of infarct size. In another study, a rat allogeneic single lung transplant model75,76 was used to compare sCR1 and sCR1-sLex, both administered prophylactically at 10 mg/kg. Assessment of graft function post-transplant showed improvements in gas exchange for both molecules compared to vehicle controls (383 mmHg, sCR1-sLex; 243 mmHg, sCR1; 56 mmHg, vehicle) with a concomitant reduction in both neutrophil migration and lipid peroxidation. As in previous studies, sCR1-sLex out-performed sCR1 in its ability to protect against tissue damage. The final rodent model described was a mouse model of acid aspiration injury in which sCR1 and sCR1-sLex were compared in both prophylactic and therapeutic settings90. Significant decreases in measured lung permeability index and polymorphonuclear cell infiltrates were observed for both molecules in prophylactic and therapeutic settings, with sCR1-sLex again showing superior efficacy compared to sCR1. The therapeutic efficacy was reduced the longer administration was delayed following induction of lung injury, and at two hours no effect was observed compared to the vehicle control90. Lastly, sCR1-sLex was tested in a NHP (baboon) model of reperfused stroke149. Although a previous study in the same NHP stroke model with unmodified sCR1 (as described above) showed no efficacy120, other studies using sCR1-sLex in a mouse model of stroke118 were successful and the authors hypothesized that adding sLex-mediated functionality to sCR1 would provide increased efficacy. Unfortunately, prophylactic administration of 15 mg/kg sCR1-sLex showed a worse outcome in NHPs than vehicle-treated animals, with a doubling of measured infarct volume at post-operative Day 3 and no change in neurological score, despite complete inhibition of complement activity for 12 hours following dosing149. The experiment was terminated following these interim results, and no further pre-clinical development of sCR1-sLex has occurred since then, given this was the last description of this molecule in the literature.

APT070 (Mirococept)

APT070 was first described150 as an N-terminal fragment of human CR1 containing only the first three SCR domains found within LHR-A, fused to a myristoylated peptide designed to bind to cell surface lipid bilayers for targeted complement inhibition at the disease site (Figure 2C). Generation of unmodified SCR1-3 began several years prior to this, when Dodd et al151 managed to successfully express and purify this protein from E. coli with yields of 6 – 15 mg/L. This and subsequent studies demonstrated that SCR1-3 was able to inhibit classical and alternative complement activity in hemolytic assays, display CFA for both C3b and C4b, and show similar DAA to the entire LHR-A domain36,151,152. However, purified SCR1-3 was still significantly less potent than soluble CR1 for both the classical/lectin and alternative pathways36. To overcome this deficiency in potency, a cysteine residue was added to the C-terminus of SCR1-3 (APT898) which allowed a membrane-localizing peptide (APT542) to be chemically coupled to it via disulphide bond formation, thereby generating APT070153. Comparative assessment of APT070 against its unmodified counterpart APT898 in human- and rat-specific assays showed cross-reactivity and significant improvements in potency (>100-fold) in vitro150,153-155.

In order to assess its efficacy in vivo, APT070 was used in several animal models of disease where complement plays a role in the indication’s pathophysiology (See Table 4 for a summary of all in vivo assessments for APT070). One of the earliest studies was in a rat model of complement-dependent acute intravascular shock, where intravenous prophylactic administration of APT070 up to 5 mg/kg showed a protective effect153,155. This was followed by a study in a model of antigen-induced arthritis154 where APT070 was administered directly to the relevant joint of rats at a 90 μg dose, resulting in a reduction in joint swelling and mean histological score compared to both vehicle control and APT898. A higher (250 μg) subsequent dose resulted in an even greater beneficial effect. A third study involved a rat kidney transplant model in which APT070 was shown to bind both glomerular endothelial and tubular epithelial cells156. Addition of APT070 to the perfusate applied to Donor’s kidneys prior to syngeneic renal transplantation led to improved graft function for up to 20 weeks. At 24 hours post-transplant, reduced acute tubular necrosis, neutrophil activity, complement deposition and blood urea nitrogen levels were observed156. Very little PK data in rats for APT070 is known with the exception of a single report stating a terminal half-life of 1 hour155, also limiting its pre-clinical use to acute settings.

APT070 then gained an additional name, Mirococept, following commercial acquisition, and was then used prophylactically in several additional animal models of disease: a mouse model of Miller-Fisher syndrome (a variant of Guillain-Barre syndrome)157; mild and severe rat intestinal IRI models158; another rat renal transplant model159; a closed-chest pig model of acute myocardial infarction160; and a transplant model whereby human pancreatic islets exposed to allogeneic whole blood were transplanted to the kidney capsule of humanized mice161. In all cases, APT070 was found to attenuate disease. APT070 was also found to be effective when administered in a therapeutic, rather than prophylactic setting. In the mouse model of Miller-Fisher syndrome, therapeutic administration of APT070 also resulted in a neuro-protective effect, although this was not as strong as observed in the prophylactic setting157. Not all animal models where APT070 was tested showed a beneficial effect. APT070 showed no effect on graft survival in a xenotransplantation model where baboons received hearts from transgenic pigs, even when administered both to the perfusate and separately to the donor itself both prior to and after reperfusion162. Similarly, in a rat model of hind limb IRI, APT070 administration had no effect on edema formation and other parameters of tissue damage163. This was in contrast to a comparator, C1-inhibitor, where a protective effect was shown. In vitro, APT070 was shown to inhibit complement activation in a cardiopulmonary bypass circuit model, and a reduction in the neutrophil activation marker CD11b was also observed164.

Table 4: Use of APT070 / Mirococept in vivo

Species

Experiment / model

Route(s) of administration

Dose(s)

Prophylactic or Therapeutic

Effecta

References

Rat

Antigen-induced Arthritis

intra-articular

90 & 250 μg/joint

Prophylactic

Protective

Linton et al (2000)154

Rat

Intravascular shock

intravenous

up to 5 mg/kg

Prophylactic

Protective

Smith and Smith (2001)153

Smith (2002)155

Rat

Kidney Allograft

Perfusion

40 μg/mL

Prophylactic

Protective

Smith and Smith (2001)153

Smith (2002)155

Pratt et al (2003)156

Mouse

Miller-Fisher Syndrome

intravenous

580 μg/mouse

Prophylactic

Protective

Halstead et al (2005)157

Mouse

Miller-Fisher Syndrome

intravenous

580 μg/mouse

Therapeutic

Protective

Halstead et al (2005)157

Rat

Intestinal IRI – mild and severe

intravenous

1, 3, 10 mg/kg

Prophylactic

Protective

Souza et al (2005)158

Rat

Kidney Allograft

Perfusion

40 μg/mL

Prophylactic

Protective

Patel et al (2006)159

Pig

Acute Myocardial Infarction

intra-coronary

0.5 mg/kg

Prophylactic

Protective

Banz et al (2007)160

Pig to Baboon

Heart Xenograft

Perfusion &

Intravenous

0.1 mg/mL (perfusion)

& 3 mg/kg (2 doses i.v.)

Prophylactic

No effect

Wu et al (2007)162

Rat

Hind Limb IRI

intravenous

9 mg/kg

Prophylactic

No effect

Duehrkop et al (2013)163

Human to Mouseb

human islet Xenograft

in vitro

0.4 μM

Prophylactic

Protective

Xiao et al (2016)161

Human

Phase I

healthy volunteers

intravenous

2, 5, 10, 20, 40, 70, 100 mg

N/A

safe and well tolerated

Smith (2002)155

Smith et al (2007)165

Kassimatis et al (2017)166

Human

Phase IIa

Kidney transplant

Perfusion

10 mg

Prophylactic

No effect

Kassimatis et al (2017)166

Human

Phase IIb

Perfusion

10 mgc

Prophylactic

No effect

Kassimatis et al (2017)166

Kassimatis et al (2021)167

Pig

Dose finding

Perfusion

20, 40, 80, 160 mg

N/A

80mg dose suitable

Kassimatis et al (2021)167

In humans, early reports of a Phase I trial of APT070 indicated that intravenous administration to healthy volunteers was safe and well-tolerated155,165. Additional information was provided in a later paper describing the design of the Phase IIb (EMPIRIKAL) trial, where 7 doses of 2, 5, 10, 20, 40, 70 and 100 mg were reported to have been administered166. Adverse events were described for the highest (100 mg) dose cohort, along with data showing the pharmacokinetics of APT070 (a plasma elimination half-life of 3 hours) and a lack of complement inhibition at doses below 10 mg166. The outcome of a pilot Phase IIa study in twelve patients was also described, where 10 mg APT070 perfused into donor kidneys pre-transplant was well tolerated with 80% of drug retained in the grafted kidney, but with no systemic complement inhibition reported and only a “trend to lower creatinine in the Mirococept group” 166. In the Phase IIb EMPIRIKAL trial which was aimed at reducing delayed graft function in transplanted kidneys, a similar (10 mg) dose of APT070 showed no efficacy, resulting in the premature termination of the study before additional doses could be tested167. In an attempt to determine a more suitable and efficacious dose of APT070 for further assessment in humans, the authors have recently conducted a dose-finding study in pigs at doses ranging from 20 to 160 mg, selecting a 80 mg dose (equivalent in humans to 120 mg) indicated as safe and sufficiently potent for further study167.

CSL040

In devising our own therapeutic candidate based on CR1, we were conscious of ensuring that any new molecule had two key benefits over its predecessors: increased potency, and improved pharmacokinetics and pharmacodynamic profiles. We were aware that there was a limit to which soluble CR1 could be truncated to create any new molecule, based on the comparatively weak complement inhibition profile of SCR1-3 compared to sCR136 and existing knowledge on the roles of the LHR domains, as described above. The strategy of adding dual functionality such as tissue/membrane targeting was not pursued, given the negative outcome of sCR1-sLex in the previously described NHP model of stroke149. So, relatively straightforward approach was devised involving the construction of a series of basic N- and C-terminal truncation variants of sCR122, which were expressed using mammalian cells and purified. Comparative assessment in complement inhibition assays quickly identified one variant, truncated at amino acid 1392 and containing the LHR-A, -B and -C domains (designated as CSL040; Figure 2D) which exhibited significantly greater in vitro potency for all three complement pathways than sCR1. Why removal of the LHR-D domain of sCR1 would produce this effect in CSL040 is not fully understood, but it is likely that a combination of increased stability and affinity for ligand, decreased steric hindrance along with the removal of interaction sites located within the LHR-D domain for C1q, MBL and certain Ficolins might all play a role22,168,169.

CSL040 was demonstrated to be a more potent inhibitor of the alternative complement pathway compared to that of the classical and lectin pathways22. This differential pathway activity can be explained mechanistically. In terms of the relative binding affinities of CR1 to C3b and C4b, 10-fold more sCR1 is required to inhibit the binding of C4b to erythrocytes compared to C3b34; a later study showed a 20-fold weaker affinity for the C4b-CR1 interaction compared to C3b-CR1170. This clearly has an effect on the DAA of CR1, with 5-10-fold more sCR1 needed to inhibit classical pathway convertase formation in vitro compared to alternative pathway convertase formation35. Other studies171,172 also showed differences in CFA for C3b compared to C4. C4b cleavage mediated by CR1 with Factor I is slower than that for C3b, with a preference for C3b if both ligands are present with Factor I. Mossakowska et al36 confirmed this finding, showing co-factor IC50 values for sCR1 to C3b of 0.8 nM compared to 15 nM for sCR1 to C4b.

Comparative assessment of CSL040 and sCR1 in a series of pharmacokinetic/ pharmacodynamic studies in both mice and rats was performed next22,173. In these studies, we took steps to ensure that the asialo-N-glycan levels of both molecules tested in these studies were similar, since previous studies showed the importance of protein sialylation for protein clearance174,175. In both species, CSL040 displayed a superior PK profile compared to sCR1. As the only point of difference between the two molecules tested, the LHR-D domain must be responsible for the faster clearance of sCR1 compared to CSL040; it was hypothesized that suboptimal glycosylation of the glycans present within LHR-D might contribute to more rapid clearance via clearance receptors such as the asialoglycoprotein or mannose receptors173. A series of additional experiments determined a relationship between the levels of CSL040 sialylation and in vivo clearance173, making this a critical quality attribute for any future in vivo studies. These experiments also demonstrated that CSL040 was safe and well tolerated at single doses of up to 90 mg/kg in both rats and non-human primates.

An analysis of the pharmacodynamic properties of CSL040 revealed an extended duration of alternative pathway inhibition, relative to the duration of the classical/lectin pathway response. An extended alternative pathway response in vivo was anticipated, given the increased in vitro potency of CSL040 for this pathway, but the extent of the response relative to the other pathways was not, since the studies performed with sCR1, sCR1-sLex, and APT070 discussed above typically only showed classical pathway activity following in vivo administration. Our data suggests some scope to potentially widen indication selection for CSL040 to chronic indications primarily involving the alternative complement pathway, rather than restricting development to acute indications.

The final studies performed to date with CSL040 have been proof-of-concept experiments in animal models of disease to evaluate in vivo efficacy. The vast body of literature for sCR1 in surrogate species (Table 1) informed decisions around indication and model selection for our own in vivo efficacy studies, and two mouse models were selected in which to test CSL040 (Table 5). The first model was a previously described176 model of immune-complex mediated kidney disease, the attenuated passive anti-glomerular basement membrane antibody-induced glomerulonephritis model, in which we found that CSL040 was able to significantly attenuate kidney damage (as measured by urine albumin output) at single 20 and 60 mg/kg doses administered prophylactically22. The second model in which CSL040 was tested was a mouse model of warm kidney IRI177. Here, we also used sCR1 (generated in-house) as a comparator, again ensuring that it retained similar asialo-N-glycan levels to that of CSL040. While two doses of 60 mg/kg CSL040 were able to significantly attenuate kidney damage, equimolar doses of 85.2 mg/kg sCR1 showed no significant effect177. This difference in relative in vivo potency is likely explained by both the 3-fold increased potency of CSL040 in vitro compared to sCR1, as well its significantly improved pharmacokinetic and pharmacodynamic properties22,173. We are now looking to expand our assessment of CSL040 to additional relevant in vivo animal models, and in settings in which CSL040 can be tested therapeutically, rather than prophylactically.

Table 5: Use of CSL040 in vivo in animal models of disease

Species

Experiment / model

Route of administration

Dose(s)

Prophylactic or Therapeutic

Effecta

References

Mouse

Glomerulonephritisb

intraperitoneal

5, 20, or 60 mg/kg

Prophylactic

Protective

Wymann et al (2021)22

Mouse

Kidney IRI

intraperitoneal

15, 30, or 60 mg/kg x 2 doses each

Prophylactic

Protective

Bongoni et al (2021)177

Summary

It has now been more than three decades since a soluble regulator of complement and potential therapeutic based on CR1 has been described. As is clear from this review, the various forms of soluble CR1 that have been engineered and developed as therapeutics since that time have shown great promise in multiple disease indications with protective effects shown in a wide variety of animal models of specific indications where complement plays a role in mediating the pathophysiology and/or progression of disease or injury. Unfortunately, it also seems clear that translating efficacy from animal models to humans for the CR1-based molecules we have reviewed has been challenging, with perhaps an over-reliance of prophylactic, rather than therapeutic animal models from which to select indications for non-human primate and human clinical studies. It is hoped that with CSL040, we can learn from past knowledge and with informed indication selection achieve sustained success in the clinic.

Supporting information

None

Conflict of interest statement

M.P.H, T.R. and S.W. are listed as inventors on International Patent Publication number WO2019/218009. All authors are CSL shareholders.

Funding

This work was wholly supported by CSL. No external sources of funding were used.

References

  1. Lachmann PJ. Biological functions of the complement system. Biochem Soc Trans. 1990; 18(6): 1143-1145.
  2. Ricklin D, Hajishengallis G, Yang K, et al. Complement: a key system for immune surveillance and homeostasis. Nat Immunol. 2010; 11(9): 785-797.
  3. Kolev M, Le Friec G, Kemper C. Complement--tapping into new sites and effector systems. Nat Rev Immunol. 2014; 14(12): 811-820.
  4. Lachmann PJ. The amplification loop of the complement pathways. Adv Immunol. 2009; 104: 115-149.
  5. Yamamoto H, Fara AF, Dasgupta P, et al. CD46: the 'multitasker' of complement proteins. Int J Biochem Cell Biol. 2013; 45(12): 2808-2820.
  6. Dho SH, Lim JC, Kim LK. Beyond the Role of CD55 as a Complement Component. Immune Netw. 2018; 18(1): e11.
  7. Davies A, Simmons DL, Hale G, et al. CD59, an LY-6-like protein expressed in human lymphoid cells, regulates the action of the complement membrane attack complex on homologous cells. J Exp Med. 1989; 170(3): 637-654.
  8. Ferreira VP, Pangburn MK, Cortes C. Complement control protein factor H: the good, the bad, and the inadequate. Mol Immunol. 2010; 47(13): 2187-2197.
  9. Lachmann PJ. The story of complement factor I. Immunobiology. 2019; 224(4): 511-517.
  10. Liu D, Niu ZX. The structure, genetic polymorphisms, expression and biological functions of complement receptor type 1 (CR1/CD35). Immunopharmacol Immunotoxicol. 2009; 31(4): 524-535.
  11. Ahearn JM, Fearon DT. Structure and function of the complement receptors, CR1 (CD35) and CR2 (CD21). Adv Immunol. 1989; 46: 183-219.
  12. Fearon DT. Identification of the membrane glycoprotein that is the C3b receptor of the human erythrocyte, polymorphonuclear leukocyte, B lymphocyte, and monocyte. J Exp Med. 1980; 152(1): 20-30.
  13. Ross GD, Lambris JD. Identification of a C3bi-specific membrane complement receptor that is expressed on lymphocytes, monocytes, neutrophils, and erythrocytes. J Exp Med. 1982; 155(1): 96-110.
  14. Karthikeyan G, Baalasubramanian S, Seth S, et al. Low levels of plasma soluble complement receptor type 1 in patients receiving thrombolytic therapy for acute myocardial infarction. J Thromb Thrombolysis. 2007; 23(2): 115-120.
  15. Hamer I, Paccaud JP, Belin D, et al. Soluble form of complement C3b/C4b receptor (CR1) results from a proteolytic cleavage in the C-terminal region of CR1 transmembrane domain. Biochem J. 1998; 329 ( Pt 1): 183-190.
  16. Krych-Goldberg M, Atkinson JP. Structure-function relationships of complement receptor type 1. Immunol Rev. 2001; 180: 112-122.
  17. Torvell M, Carpanini SM, Daskoulidou N, et al. Genetic Insights into the Impact of Complement in Alzheimer's Disease. Genes (Basel). 2021; 12(12).
  18. Weis JH, Morton CC, Bruns GA, et al. A complement receptor locus: genes encoding C3b/C4b receptor and C3d/Epstein-Barr virus receptor map to 1q32. J Immunol. 1987; 138(1): 312-315.
  19. Klickstein LB, Wong WW, Smith JA, et al. Human C3b/C4b receptor (CR1). Demonstration of long homologous repeating domains that are composed of the short consensus repeats characteristics of C3/C4 binding proteins. J Exp Med. 1987; 165(4): 1095-1112.
  20. Kalli KR, Hsu PH, Bartow TJ, et al. Mapping of the C3b-binding site of CR1 and construction of a (CR1)2-F(ab')2 chimeric complement inhibitor. J Exp Med. 1991; 174(6): 1451-1460.
  21. Mqadmi A, Abdullah Y, Yazdanbakhsh K. Characterization of complement receptor 1 domains for prevention of complement-mediated red cell destruction. Transfusion. 2005; 45(2): 234-244.
  22. Wymann S, Dai Y, Nair AG, et al. A novel soluble complement receptor 1 fragment with enhanced therapeutic potential. J Biol Chem. 2021; 296: 100200.
  23. Kirkitadze MD, Barlow PN. Structure and flexibility of the multiple domain proteins that regulate complement activation. Immunol Rev. 2001; 180: 146-161.
  24. Weisman HF, Bartow T, Leppo MK, et al. Recombinant soluble CR1 suppressed complement activation, inflammation, and necrosis associated with reperfusion of ischemic myocardium. Trans Assoc Am Physicians. 1990; 103: 64-72.
  25. Gilbert HE, Asokan R, Holers VM, et al. The 15 SCR flexible extracellular domains of human complement receptor type 2 can mediate multiple ligand and antigen interactions. J Mol Biol. 2006; 362(5): 1132-1147.
  26. Furtado PB, Huang CY, Ihyembe D, et al. The partly folded back solution structure arrangement of the 30 SCR domains in human complement receptor type 1 (CR1) permits access to its C3b and C4b ligands. J Mol Biol. 2008; 375(1): 102-118.
  27. Lublin DM, Griffith RC, Atkinson JP. Influence of glycosylation on allelic and cell-specific Mr variation, receptor processing, and ligand binding of the human complement C3b/C4b receptor. J Biol Chem. 1986; 261(13): 5736-5744.
  28. Ross GD. Analysis of the different types of leukocyte membrane complement receptors and their interaction with the complement system. J Immunol Methods. 1980; 37(3-4): 197-211.
  29. Fearon DT. Regulation of the amplification C3 convertase of human complement by an inhibitory protein isolated from human erythrocyte membrane. Proc Natl Acad Sci U S A. 1979; 76(11): 5867-5871.
  30. Iida K, Nussenzweig V. Complement receptor is an inhibitor of the complement cascade. J Exp Med. 1981; 153(5): 1138-1150.
  31. Medof ME, Iida K, Mold C, et al. Unique role of the complement receptor CR1 in the degradation of C3b associated with immune complexes. J Exp Med. 1982; 156(6): 1739-1754.
  32. Ross GD, Lambris JD, Cain JA, et al. Generation of three different fragments of bound C3 with purified factor I or serum. I. Requirements for factor H vs CR1 cofactor activity. J Immunol. 1982; 129(5): 2051-2060.
  33. Alcorlo M, Martinez-Barricarte R, Fernandez FJ, et al. Unique structure of iC3b resolved at a resolution of 24 A by 3D-electron microscopy. Proc Natl Acad Sci U S A. 2011; 108(32): 13236-13240.
  34. Weisman HF, Bartow T, Leppo MK, et al. Soluble human complement receptor type 1: in vivo inhibitor of complement suppressing post-ischemic myocardial inflammation and necrosis. Science. 1990; 249(4965): 146-151.
  35. Wong WW, Farrell SA. Proposed structure of the F' allotype of human CR1. Loss of a C3b binding site may be associated with altered function. J Immunol. 1991; 146(2): 656-662.
  36. Mossakowska D, Dodd I, Pindar W, et al. Structure-activity relationships within the N-terminal short consensus repeats (SCR) of human CR1 (C3b/C4b receptor, CD35): SCR 3 plays a critical role in inhibition of the classical and alternative pathways of complement activation. Eur J Immunol. 1999; 29(6): 1955-1965.
  37. Pratt JR, Hibbs MJ, Laver AJ, et al. Effects of complement inhibition with soluble complement receptor-1 on vascular injury and inflammation during renal allograft rejection in the rat. Am J Pathol. 1996; 149(6): 2055-2066.
  38. Makrides SC, Nygren PA, Andrews B, et al. Extended in vivo half-life of human soluble complement receptor type 1 fused to a serum albumin-binding receptor. J Pharmacol Exp Ther. 1996; 277(1): 534-542.
  39. Ong GL, Mattes MJ. Mouse strains with typical mammalian levels of complement activity. J Immunol Methods. 1989; 125(1-2): 147-158.
  40. Cheung AK, Parker CJ, Hohnholt M. Soluble complement receptor type 1 inhibits complement activation induced by hemodialysis membranes in vitro. Kidney Int. 1994; 46(6): 1680-1687.
  41. Himmelfarb J, McMonagle E, Holbrook D, et al. Soluble complement receptor 1 inhibits both complement and granulocyte activation during ex vivo hemodialysis. J Lab Clin Med. 1995; 126(4): 392-400.
  42. Hill J, Lindsay TF, Ortiz F, et al. Soluble complement receptor type 1 ameliorates the local and remote organ injury after intestinal ischemia-reperfusion in the rat. J Immunol. 1992; 149(5): 1723-1728.
  43. Lindsay TF, Hill J, Ortiz F, et al. Blockade of complement activation prevents local and pulmonary albumin leak after lower torso ischemia-reperfusion. Ann Surg. 1992; 216(6): 677-683.
  44. Pemberton M, Anderson G, Vetvicka V, et al. Microvascular effects of complement blockade with soluble recombinant CR1 on ischemia/reperfusion injury of skeletal muscle. J Immunol. 1993; 150(11): 5104-5113.
  45. Shandelya SM, Kuppusamy P, Herskowitz A, et al. Soluble complement receptor type 1 inhibits the complement pathway and prevents contractile failure in the postischemic heart. Evidence that complement activation is required for neutrophil-mediated reperfusion injury. Circulation. 1993; 88(6): 2812-2826.
  46. Smith EF, 3rd, Griswold DE, Egan JW, et al. Reduction of myocardial reperfusion injury with human soluble complement receptor type 1 (BRL 55730). Eur J Pharmacol. 1993; 236(3): 477-481.
  47. Chavez-Cartaya RE, DeSola GP, Wright L, et al. Regulation of the complement cascade by soluble complement receptor type 1. Protective effect in experimental liver ischemia and reperfusion. Transplantation. 1995; 59(7): 1047-1052.
  48. Xiao F, Eppihimer MJ, Willis BH, et al. Complement-mediated lung injury and neutrophil retention after intestinal ischemia-reperfusion. J Appl Physiol (1985). 1997; 82(5): 1459-1465.
  49. Austen WG, Jr., Kyriakides C, Favuzza J, et al. Intestinal ischemia-reperfusion injury is mediated by the membrane attack complex. Surgery. 1999; 126(2): 343-348.
  50. Eror AT, Stojadinovic A, Starnes BW, et al. Antiinflammatory effects of soluble complement receptor type 1 promote rapid recovery of ischemia/reperfusion injury in rat small intestine. Clin Immunol. 1999; 90(2): 266-275.
  51. Williams JP, Pechet TT, Weiser MR, et al. Intestinal reperfusion injury is mediated by IgM and complement. J Appl Physiol (1985). 1999; 86(3): 938-942.
  52. Zacharowski K, Otto M, Hafner G, et al. Thiemermann C. Reduction of myocardial infarct size with sCR1sLe(x), an alternatively glycosylated form of human soluble complement receptor type 1 (sCR1), possessing sialyl Lewis x. Br J Pharmacol. 1999; 128(5): 945-952.
  53. Lehmann TG, Koeppel TA, Munch S, et al. Impact of inhibition of complement by sCR1 on hepatic microcirculation after warm ischemia. Microvasc Res. 2001; 62(3): 284-292.
  54. von Dobschuetz E, Bleiziffer O, Pahernik S, et al. Soluble complement receptor 1 preserves endothelial barrier function and microcirculation in postischemic pancreatitis in the rat. Am J Physiol Gastrointest Liver Physiol. 2004; 286(5): G791-796.
  55. Lillegard KE, Johnson AC, Lojovich SJ, et al. Complement activation is critical for placental ischemia-induced hypertension in the rat. Mol Immunol. 2013; 56(1-2): 91-97.
  56. Regal JF, Lund JM, Wing CR, et al. Interactions between the complement and endothelin systems in normal pregnancy and following placental ischemia. Mol Immunol. 2019; 114: 10-18.
  57. Hameed AM, Lu DB, Burns H, et al. Pharmacologic targeting of renal ischemia-reperfusion injury using a normothermic machine perfusion platform. Sci Rep. 2020; 10(1): 6930.
  58. Pruitt SK, Baldwin WM, 3rd, Marsh HC, Jr., et al. The effect of soluble complement receptor type 1 on hyperacute xenograft rejection. Transplantation. 1991; 52(5): 868-873.
  59. Chrupcala M, Pomer S, Staehler G, et al. Prolongation of discordant renal xenograft survival by depletion of complement. Comparative effects of systemically administered cobra venom factor and soluble complement receptor type 1 in a guinea-pig to rat model. Transpl Int. 1994; 7 Suppl 1: S650-653.
  60. Zehr KJ, Herskowitz A, Lee PC, et al. Neutrophil adhesion and complement inhibition prolongs survival of cardiac xenografts in discordant species. Transplantation. 1994; 57(6): 900-906.
  61. Pruitt SK, Kirk AD, Bollinger RR, et al. The effect of soluble complement receptor type 1 on hyperacute rejection of porcine xenografts. Transplantation. 1994; 57(3): 363-370.
  62. Candinas D, Lesnikoski BA, Robson SC, et al. Effect of repetitive high-dose treatment with soluble complement receptor type 1 and cobra venom factor on discordant xenograft survival. Transplantation. 1996; 62(3): 336-342.
  63. Davis EA, Pruitt SK, Greene PS, et al. Inhibition of complement, evoked antibody, and cellular response prevents rejection of pig-to-primate cardiac xenografts. Transplantation. 1996; 62(7): 1018-1023.
  64. Pratt JR, Hibbs MJ, Laver AJ, et al. Allograft immune response with sCR1 intervention. Transpl Immunol. 1996; 4(1): 72-75.
  65. Pratt JR, Harmer AW, Levin J, et al. Influence of complement on the allospecific antibody response to a primary vascularized organ graft. Eur J Immunol. 1997; 27(11): 2848-2853.
  66. Fujiwara I, Nakajima H, Akioka K, et al. Soluble complement receptor type 1 and antithrombin-III combination therapy prolongs xenograft survival: the role of thrombin and prostacyclin in hyperacute rejection. Transplant Proc. 1997; 29(1-2): 935-937.
  67. Naka Y, Marsh HC, Scesney SM, et al. Complement activation as a cause for primary graft failure in an isogeneic rat model of hypothermic lung preservation and transplantation. Transplantation. 1997; 64(9): 1248-1255.
  68. Pruitt SK, Bollinger RR, Collins BH, et al. Effect of continuous complement inhibition using soluble complement receptor type 1 on survival of pig-to-primate cardiac xenografts. Transplantation. 1997; 63(6): 900-902.
  69. Lehmann TG, Koeppel TA, Kirschfink M, et al. Complement inhibition by soluble complement receptor type 1 improves microcirculation after rat liver transplantation. Transplantation. 1998; 66(6): 717-722.
  70. Pierre AF, Xavier AM, Liu M, et al. Effect of complement inhibition with soluble complement receptor 1 on pig allotransplant lung function. Transplantation. 1998; 66(6): 723-732.
  71. Schmid RA, Zollinger A, Singer T, et al. Effect of soluble complement receptor type 1 on reperfusion edema and neutrophil migration after lung allotransplantation in swine. J Thorac Cardiovasc Surg. 1998; 116(1): 90-97.
  72. Bennet W, Sundberg B, Lundgren T, et al. Damage to porcine islets of Langerhans after exposure to human blood in vitro, or after intraportal transplantation to cynomologus monkeys: protective effects of sCR1 and heparin. Transplantation. 2000; 69(5): 711-719.
  73. Lundgren T, Bennet W, Tibell A, et al. Soluble complement receptor 1 (TP10) preserves adult porcine islet morphology after intraportal transplantation into cynomolgus monkeys. Transplant Proc. 2001; 33(1-2): 725.
  74. Kallio EA, Lemstrom KB, Hayry PJ, et al. Blockade of complement inhibits obliterative bronchiolitis in rat tracheal allografts. Am J Respir Crit Care Med. 2000; 161(4 Pt 1): 1332-1339.
  75. Stammberger U, Hamacher J, Hillinger S, et al. sCR1sLe ameliorates ischemia/reperfusion injury in experimental lung transplantation. J Thorac Cardiovasc Surg. 2000; 120(6): 1078-1084.
  76. Schmid RA, Hillinger S, Hamacher J, et al. TP20 is superior to TP10 in reducing ischemia/reperfusion injury in rat lung grafts. Transplant Proc. 2001; 33(1-2): 948-949.
  77. Lam TT, Hausen B, Hook L, et al. The effect of soluble complement receptor type 1 on acute humoral xenograft rejection in hDAF-transgenic pig-to-primate life-supporting kidney xenografts. Xenotransplantation. 2005; 12(1): 20-29.
  78. Damman J, Hoeger S, Boneschansker L, et al. Targeting complement activation in brain-dead donors improves renal function after transplantation. Transpl Immunol. 2011; 24(4): 233-237.
  79. Mulligan MS, Yeh CG, Rudolph AR, et al. Protective effects of soluble CR1 in complement- and neutrophil-mediated tissue injury. J Immunol. 1992; 148(5): 1479-1485.
  80. Rabinovici R, Yeh CG, Hillegass LM, et al. Role of complement in endotoxin/platelet-activating factor-induced lung injury. J Immunol. 1992; 149(5): 1744-1750.
  81. Homeister JW, Satoh PS, Kilgore KS, et al. Soluble complement receptor type 1 prevents human complement-mediated damage of the rabbit isolated heart. J Immunol. 1993; 150(3): 1055-1064.
  82. Rabinovici R, Sofronski MD, Borboroglu P, et al. Interleukin-2-induced lung injury. The role of complement. Circ Res. 1994; 74(2): 329-335.
  83. Gralinski MR, Wiater BC, Assenmacher AN, et al. Selective inhibition of the alternative complement pathway by sCR1[desLHR-A] protects the rabbit isolated heart from human complement-mediated damage. Immunopharmacology. 1996; 34(2-3): 79-88.
  84. Nishizawa H, Yamada H, Miyazaki H, et al. Soluble complement receptor type 1 inhibited the systemic organ injury caused by acid instillation into a lung. Anesthesiology. 1996; 85(5): 1120-1128.
  85. Weiser MR, Pechet TT, Williams JP, et al. Experimental murine acid aspiration injury is mediated by neutrophils and the alternative complement pathway. J Appl Physiol (1985). 1997; 83(4): 1090-1095.
  86. Nomura N, Lim HW, Levin JL, et al. Effect of soluble complement receptor type 1 on porphyrin-induced phototoxicity in guinea pigs. J Photochem Photobiol B. 1998; 42(1): 28-31.
  87. Mulligan MS, Warner RL, Rittershaus CW, et al. Endothelial targeting and enhanced antiinflammatory effects of complement inhibitors possessing sialyl Lewisx moieties. J Immunol. 1999; 162(8): 4952-4959.
  88. Heller A, Kunz M, Samakas A, et al. The complement regulators C1 inhibitor and soluble complement receptor 1 attenuate acute lung injury in rabbits. Shock. 2000; 13(4): 285-290.
  89. Tanhehco EJ, Lee H, Lucchesi BR. Sublytic complement attack reduces infarct size in rabbit isolated hearts: evidence for C5a-mediated cardioprotection. Immunopharmacology. 2000; 49(3): 391-399.
  90. Kyriakides C, Wang Y, Austen WG, Jr., et al. Sialyl Lewis(x) hybridized complement receptor type 1 moderates acid aspiration injury. Am J Physiol Lung Cell Mol Physiol. 2001; 281(6): L1494-1499.
  91. Azimzadeh A, Zorn GL, 3rd, Blair KS, et al. Hyperacute lung rejection in the pig-to-human model. 2. Synergy between soluble and membrane complement inhibition. Xenotransplantation. 2003; 10(2): 120-131.
  92. Pfeiffer S, Zorn GL, 3rd, Zhang JP, et al. Hyperacute lung rejection in the pig-to-human model. III. Platelet receptor inhibitors synergistically modulate complement activation and lung injury. Transplantation. 2003; 75(7): 953-959.
  93. Schroder C, Wu GS, Price E, et al. Hyperacute rejection of mouse lung by human blood: characterization of the model and the role of complement. Transplantation. 2003; 76(5): 755-760.
  94. Regal JF, Laule CF, McCutcheon L, et al. The complement system in hypertension and renal damage in the Dahl SS rat. Physiol Rep. 2018; 6(6): e13655.
  95. Yeh CG, Marsh HC, Jr., Carson GR, et al. Recombinant soluble human complement receptor type 1 inhibits inflammation in the reversed passive arthus reaction in rats. J Immunol. 1991; 146(1): 250-256.
  96. Fava RA, Gates C, Townes AS. Critical role of peripheral blood phagocytes and the involvement of complement in tumour necrosis factor enhancement of passive collagen-arthritis. Clin Exp Immunol. 1993; 94(2): 261-266.
  97. Couser WG, Johnson RJ, Young BA, et al. The effects of soluble recombinant complement receptor 1 on complement-mediated experimental glomerulonephritis. J Am Soc Nephrol. 1995; 5(11): 1888-1894.
  98. Metcalfe RA, McIntosh RS, Morgan BP, et al. The effect of soluble complement receptor 1 (sCR1) and human thyroid antibodies on the course of experimental autoimmune thyroiditis in rats. Autoimmunity. 1996; 23(1): 1-8.
  99. Weiser MR, Gibbs SA, Moore FD, Jr., et al. Complement inhibition by soluble complement receptor type 1 fails to moderate cerulein-induced pancreatitis in the rat. Int J Pancreatol. 1996; 19(2): 129-134.
  100. Acioli JM, Isobe M, Kawasaki S. Early complement system activation and neutrophil priming in acute pancreatitis: participation of trypsin. Surgery. 1997; 122(5): 909-917.
  101. Goodfellow RM, Williams AS, Levin JL, et al. Local therapy with soluble complement receptor 1 (sCR1) suppresses inflammation in rat mono-articular arthritis. Clin Exp Immunol. 1997; 110(1): 45-52.
  102. Lima MC, Prouvost-Danon A, e Silva PM, et al. Studies on the mechanisms involved in antigen-evoked pleural inflammation in rats: contribution of IgE and complement. J Leukoc Biol. 1997; 61(3): 286-292.
  103. Frenette J, Cai B, Tidball JG. Complement activation promotes muscle inflammation during modified muscle use. Am J Pathol. 2000; 156(6): 2103-2110.
  104. Goodfellow RM, Williams AS, Levin JL, et al. Soluble complement receptor one (sCR1) inhibits the development and progression of rat collagen-induced arthritis. Clin Exp Immunol. 2000; 119(1): 210-216.
  105. Mizuno M, Nishikawa K, Morgan BP, et al. Comparison of the suppressive effects of soluble CR1 and C5a receptor antagonist in acute arthritis induced in rats by blocking of CD59. Clin Exp Immunol. 2000; 119(2): 368-375.
  106. Abe M, Shibata K, Akatsu H, et al. Contribution of anaphylatoxin C5a to late airway responses after repeated exposure of antigen to allergic rats. J Immunol. 2001; 167(8): 4651-4660.
  107. Kondo C, Mizuno M, Nishikawa K, et al. The role of C5a in the development of thrombotic glomerulonephritis in rats. Clin Exp Immunol. 2001; 124(2): 323-329.
  108. Hartwig W, Klafs M, Kirschfink M, et al. Interaction of complement and leukocytes in severe acute pancreatitis: potential for therapeutic intervention. Am J Physiol Gastrointest Liver Physiol. 2006; 291(5): G844-850.
  109. Zhang Y, Nester CM, Holanda DG, et al. Soluble CR1 therapy improves complement regulation in C3 glomerulopathy. J Am Soc Nephrol. 2013; 24(11): 1820-1829.
  110. Gillinov AM, DeValeria PA, Winkelstein JA, et al. Complement inhibition with soluble complement receptor type 1 in cardiopulmonary bypass. Ann Thorac Surg. 1993; 55(3): 619-624.
  111. Lazar HL, Bao Y, Gaudiani J, et al. Total complement inhibition: an effective strategy to limit ischemic injury during coronary revascularization on cardiopulmonary bypass. Circulation. 1999; 100(13): 1438-1442.
  112. Chai PJ, Nassar R, Oakeley AE, et al. Soluble complement receptor-1 protects heart, lung, and cardiac myofilament function from cardiopulmonary bypass damage. Circulation. 2000; 101(5): 541-546.
  113. Piddlesden SJ, Storch MK, Hibbs M, et al. Soluble recombinant complement receptor 1 inhibits inflammation and demyelination in antibody-mediated demyelinating experimental allergic encephalomyelitis. J Immunol. 1994; 152(11): 5477-5484.
  114. Jung S, Toyka KV, Hartung HP. Soluble complement receptor type 1 inhibits experimental autoimmune neuritis in Lewis rats. Neurosci Lett. 1995; 200(3): 167-170.
  115. Kaczorowski SL, Schiding JK, Toth CA, et al. Effect of soluble complement receptor-1 on neutrophil accumulation after traumatic brain injury in rats. J Cereb Blood Flow Metab. 1995; 15(5): 860-864.
  116. Piddlesden SJ, Jiang S, Levin JL, et al. Soluble complement receptor 1 (sCR1) protects against experimental autoimmune myasthenia gravis. J Neuroimmunol. 1996; 71(1-2): 173-177.
  117. Vriesendorp FJ, Flynn RE, Pappolla MA, et al. Soluble complement receptor 1 (sCR1) is not as effective as cobra venom factor in the treatment of experimental allergic neuritis. Int J Neurosci. 1997; 92(3-4): 287-298.
  118. Huang J, Kim LJ, Mealey R, et al. Neuronal protection in stroke by an sLex-glycosylated complement inhibitory protein. Science. 1999; 285(5427): 595-599.
  119. Twining CM, Sloane EM, Schoeniger DK, et al. Activation of the spinal cord complement cascade might contribute to mechanical allodynia induced by three animal models of spinal sensitization. J Pain. 2005; 6(3): 174-183.
  120. Mocco J, Mack WJ, Ducruet AF, et al. Preclinical evaluation of the neuroprotective effect of soluble complement receptor type 1 in a nonhuman primate model of reperfused stroke. J Neurosurg. 2006; 105(4): 595-601.
  121. Ramaglia V, Wolterman R, de Kok M, et al. Soluble complement receptor 1 protects the peripheral nerve from early axon loss after injury. Am J Pathol. 2008; 172(4): 1043-1052.
  122. Ramaglia V, Tannemaat MR, de Kok M, et al. Complement inhibition accelerates regeneration in a model of peripheral nerve injury. Mol Immunol. 2009; 47(2-3): 302-309.
  123. Li LM, Li JB, Zhu Y, et al. Soluble complement receptor type 1 inhibits complement system activation and improves motor function in acute spinal cord injury. Spinal Cord. 2010; 48(2): 105-111.
  124. Regal JF, Fraser DG, Toth CA. Role of the complement system in antigen-induced bronchoconstriction and changes in blood pressure in the guinea pig. J Pharmacol Exp Ther. 1993; 267(2): 979-988.
  125. Swift AJ, Collins TS, Bugelski P, et al. Soluble human complement receptor type 1 inhibits complement-mediated host defense. Clin Diagn Lab Immunol. 1994; 1(5): 585-589.
  126. Fruchterman TM, Spain DA, Wilson MA, et al. Complement inhibition prevents gut ischemia and endothelial cell dysfunction after hemorrhage/resuscitation. Surgery. 1998; 124(4): 782-791; discussion 791-782.
  127. Spain DA, Fruchterman TM, Matheson PJ, et al. Complement activation mediates intestinal injury after resuscitation from hemorrhagic shock. J Trauma. 1999; 46(2): 224-233.
  128. Wagner E, Platt JL, Howell DN, et al. IgG and complement-mediated tissue damage in the absence of C2: evidence of a functionally active C2-bypass pathway in a guinea pig model. J Immunol. 1999; 163(6): 3549-3558.
  129. Mizuno M, Nishikawa K, Okada N, et al. Soluble complement receptor type 1 protects rats from lethal shock induced by anti-Crry antibody following lipopolysaccharide priming. Int Arch Allergy Immunol. 2002; 127(1): 55-62.
  130. Yazdanbakhsh K, Kang S, Tamasauskas D, et al. Complement receptor 1 inhibitors for prevention of immune-mediated red cell destruction: potential use in transfusion therapy. Blood. 2003; 101(12): 5046-5052.
  131. Chen D, Song MQ, Liu YJ, et al. Inhibition of complement C3 might rescue vascular hyporeactivity in a conscious hemorrhagic shock rat model. Microvasc Res. 2016; 105: 23-29.
  132. Zimmerman JL, Dellinger RP, Straube RC, et al. Phase I trial of the recombinant soluble complement receptor 1 in acute lung injury and acute respiratory distress syndrome. Crit Care Med. 2000; 28(9): 3149-3154.
  133. Li JS, Sanders SP, Perry AE, et al. Pharmacokinetics and safety of TP10, soluble complement receptor 1, in infants undergoing cardiopulmonary bypass. Am Heart J. 2004; 147(1): 173-180.
  134. Lazar HL, Bokesch PM, van Lenta F, et al. Soluble human complement receptor 1 limits ischemic damage in cardiac surgery patients at high risk requiring cardiopulmonary bypass. Circulation. 2004; 110(11 Suppl 1): II274-279.
  135. Lazar HL, Keilani T, Fitzgerald CA, et al. Beneficial effects of complement inhibition with soluble complement receptor 1 (TP10) during cardiac surgery: is there a gender difference? Circulation. 2007; 116(11 Suppl): I83-88.
  136. Pierson RN, 3rd, Loyd JE, Goodwin A, et al. Successful management of an ABO-mismatched lung allograft using antigen-specific immunoadsorption, complement inhibition, and immunomodulatory therapy. Transplantation. 2002; 74(1): 79-84.
  137. Keshavjee S, Davis RD, Zamora MR, et al. A randomized, placebo-controlled trial of complement inhibition in ischemia-reperfusion injury after lung transplantation in human beings. J Thorac Cardiovasc Surg. 2005; 129(2): 423-428.
  138. Ricklin D, Mastellos DC, Reis ES, et al. The renaissance of complement therapeutics. Nat Rev Nephrol. 2018; 14(1): 26-47.
  139. Dicker M, Strasser R. Using glyco-engineering to produce therapeutic proteins. Expert Opin Biol Ther. 2015; 15(10): 1501-1516.
  140. Rosen SD, Bertozzi CR. The selectins and their ligands. Curr Opin Cell Biol. 1994; 6(5): 663-673.
  141. Foxall C, Watson SR, Dowbenko D, et al. The three members of the selectin receptor family recognize a common carbohydrate epitope, the sialyl Lewis(x) oligosaccharide. J Cell Biol. 1992; 117(4): 895-902.
  142. Lasky LA. Selectins: interpreters of cell-specific carbohydrate information during inflammation. Science. 1992; 258(5084): 964-969.
  143. Gonzalez-Amaro R, Diaz-Gonzalez F, Sanchez-Madrid F. Adhesion molecules in inflammatory diseases. Drugs. 1998; 56(6): 977-988.
  144. Rittershaus CW, Thomas LJ, Miller DP, et al. Recombinant glycoproteins that inhibit complement activation and also bind the selectin adhesion molecules. J Biol Chem. 1999; 274(16): 11237-11244.
  145. Mulligan MS, Lowe JB, Larsen RD, et al. Protective effects of sialylated oligosaccharides in immune complex-induced acute lung injury. J Exp Med. 1993; 178(2): 623-631.
  146. Mulligan MS, Paulson JC, De Frees S, et al. Protective effects of oligosaccharides in P-selectin-dependent lung injury. Nature. 1993; 364(6433): 149-151.
  147. Stanley P, Atkinson PH. The LEC11 Chinese hamster ovary mutant synthesizes N-linked carbohydrates containing sialylated, fucosylated lactosamine units. Analysis by one- and two-dimensional 1H NMR spectroscopy. J Biol Chem. 1988; 263(23): 11374-11381.
  148. Picard MD, Pettey CL, Marsh HC, et al. Characterization of N-linked oligosaccharides bearing sialyl lewis x moieties on an alternatively glycosylated form of soluble complement receptor type 1 (sCR1). Biotechnol Appl Biochem. 2000; 31(1): 5-13.
  149. Ducruet AF, Mocco J, Mack WJ, et al. Pre-clinical evaluation of an sLe x-glycosylated complement inhibitory protein in a non-human primate model of reperfused stroke. J Med Primatol. 2007; 36(6): 375-380.
  150. Dong J, Pratt JR, Smith RA, et al. Strategies for targeting complement inhibitors in ischaemia/reperfusion injury. Mol Immunol. 1999; 36(13-14): 957-963.
  151. Dodd I, Mossakowska DE, Camilleri P, et al. Overexpression in Escherichia coli, folding, purification, and characterization of the first three short consensus repeat modules of human complement receptor type 1. Protein Expr Purif. 1995; 6(6): 727-736.
  152. Krych-Goldberg M, Hauhart RE, Subramanian VB, et al. Decay accelerating activity of complement receptor type 1 (CD35). Two active sites are required for dissociating C5 convertases. J Biol Chem. 1999; 274(44): 31160-31168.
  153. Smith GP, Smith RA. Membrane-targeted complement inhibitors. Mol Immunol. 2001; 38(2-3): 249-255.
  154. Linton SM, Williams AS, Dodd I, et al. Therapeutic efficacy of a novel membrane-targeted complement regulator in antigen-induced arthritis in the rat. Arthritis Rheum. 2000; 43(11): 2590-2597.
  155. Smith RA. Targeting anticomplement agents. Biochem Soc Trans. 2002; 30(Pt 6): 1037-1041.
  156. Pratt JR, Jones ME, Dong J, et al. Nontransgenic hyperexpression of a complement regulator in donor kidney modulates transplant ischemia/reperfusion damage, acute rejection, and chronic nephropathy. Am J Pathol. 2003; 163(4): 1457-1465.
  157. Halstead SK, Humphreys PD, Goodfellow JA, et al. Complement inhibition abrogates nerve terminal injury in Miller Fisher syndrome. Ann Neurol. 2005; 58(2): 203-210.
  158. Souza DG, Esser D, Bradford R, et al. APT070 (Mirococept), a membrane-localised complement inhibitor, inhibits inflammatory responses that follow intestinal ischaemia and reperfusion injury. Br J Pharmacol. 2005; 145(8): 1027-1034.
  159. Patel H, Smith RA, Sacks SH, et al. Therapeutic strategy with a membrane-localizing complement regulator to increase the number of usable donor organs after prolonged cold storage. J Am Soc Nephrol. 2006; 17(4): 1102-1111.
  160. Banz Y, Hess OM, Robson SC, et al. Attenuation of myocardial reperfusion injury in pigs by Mirococept, a membrane-targeted complement inhibitor derived from human CR1. Cardiovasc Res. 2007; 76(3): 482-493.
  161. Xiao F, Ma L, Zhao M, et al. APT070 (mirococept), a membrane-localizing C3 convertase inhibitor, attenuates early human islet allograft damage in vitro and in vivo in a humanized mouse model. Br J Pharmacol. 2016; 173(3): 575-587.
  162. Wu G, Pfeiffer S, Schroder C, et al. Coagulation cascade activation triggers early failure of pig hearts expressing human complement regulatory genes. Xenotransplantation. 2007; 14(1): 34-47.
  163. Duehrkop C, Banz Y, Spirig R, et al. C1 esterase inhibitor reduces lower extremity ischemia/reperfusion injury and associated lung damage. PLoS One. 2013; 8(8): e72059.
  164. De Silva RJ, Vuylsteke A, Fritchley SJ, et al. APT070 inhibits complement activation during in vitro cardiopulmonary bypass. Eur J Cardiothorac Surg. 2006; 30(1): 72-76.
  165. Proceedings and abstracts of the XIth European Meeting on Complement in Human Disease. Mol Immunol. 2007; 44(16): 3809-3994.
  166. Kassimatis T, Qasem A, Douiri A, et al. A double-blind randomised controlled investigation into the efficacy of Mirococept (APT070) for preventing ischaemia reperfusion injury in the kidney allograft (EMPIRIKAL): study protocol for a randomised controlled trial. Trials. 2017; 18(1): 255.
  167. Kassimatis T, Greenlaw R, Hunter JP, et al. Ex vivo delivery of Mirococept: A dose-finding study in pig kidney after showing a low dose is insufficient to reduce delayed graft function in human kidney. Am J Transplant. 2021; 21(3): 1012-1026.
  168. Klickstein LB, Barbashov SF, Liu T, et al. Complement receptor type 1 (CR1, CD35) is a receptor for C1q. Immunity. 1997; 7(3): 345-355.
  169. Jacquet M, Lacroix M, Ancelet S, et al. Deciphering complement receptor type 1 interactions with recognition proteins of the lectin complement pathway. J Immunol. 2013; 190(7): 3721-3731.
  170. Reilly BD, Makrides SC, Ford PJ, et al. Quantitative analysis of C4b dimer binding to distinct sites on the C3b/C4b receptor (CR1). J Biol Chem. 1994; 269(10): 7696-7701.
  171. Medof ME, Nussenzweig V. Control of the function of substrate-bound C4b-C3b by the complement receptor Cr1. J Exp Med. 1984; 159(6): 1669-1685.
  172. Krych M, Clemenza L, Howdeshell D, et al. Analysis of the functional domains of complement receptor type 1 (C3b/C4b receptor; CD35) by substitution mutagenesis. J Biol Chem. 1994; 269(18): 13273-13278.
  173. Wymann S, Mischnik M, Leong D, et al. Sialylation-dependent pharmacokinetics and differential complement pathway inhibition are hallmarks of CR1 activity in vivo. Biochem J. 2022; 479(9): 1007-1030.
  174. Morell AG, Gregoriadis G, Scheinberg IH, et al. The role of sialic acid in determining the survival of glycoproteins in the circulation. J Biol Chem. 1971; 246(5): 1461-1467.
  175. Park EI, Manzella SM, Baenziger JU. Rapid clearance of sialylated glycoproteins by the asialoglycoprotein receptor. J Biol Chem. 2003; 278(7): 4597-4602.
  176. Otten MA, Groeneveld TW, Flierman R, et al. Both complement and IgG fc receptors are required for development of attenuated antiglomerular basement membrane nephritis in mice. J Immunol. 2009; 183(6): 3980-3988.
  177. Bongoni AK, Vikstrom IB, McRae JL, et al. A potent truncated form of human soluble CR1 is protective in a mouse model of renal ischemia-reperfusion injury. Sci Rep. 2021; 11(1): 21873.
 

Article Info

Article Notes

  • Published on: October 21, 2022

Keywords

  • Soluble
  • Complement
  • Receptor
  • Therapeutic
  • CSL040
  • Model
  • Recombinant
  • TP10

*Correspondence:

Dr. Matthew P. Hardy,
CSL, Bio21 Institute, Victoria, Australia;
Email: Matt.Hardy@csl.com.au

Copyright: ©2022 Hardy MP. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License.