WO2016193402A1 - Pseudomonas vaccine - Google Patents
Pseudomonas vaccine Download PDFInfo
- Publication number
- WO2016193402A1 WO2016193402A1 PCT/EP2016/062574 EP2016062574W WO2016193402A1 WO 2016193402 A1 WO2016193402 A1 WO 2016193402A1 EP 2016062574 W EP2016062574 W EP 2016062574W WO 2016193402 A1 WO2016193402 A1 WO 2016193402A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- oprf
- subject
- disorder
- antigen
- hepatobiliary
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Ceased
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/02—Bacterial antigens
- A61K39/104—Pseudomonadales, e.g. Pseudomonas
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/195—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
- C07K14/21—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Pseudomonadaceae (F)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/12—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
- C07K16/1203—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria
- C07K16/1214—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-negative bacteria from Pseudomonadaceae (F)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/01—Fusion polypeptide containing a localisation/targetting motif
- C07K2319/02—Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/20—Fusion polypeptide containing a tag with affinity for a non-protein ligand
- C07K2319/21—Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
Definitions
- the disclosure relates to methods for the reduction of mortality and/or the treatment and/or prevention of Pseudomonas aeruginosa infection in subjects with hepatobiliary disorders and/or in patients with no respiratory, thoracic or mediastinal disorders.
- Nosocomial infections, or hospital-acquired infections are a significant cause of morbidity and mortality in both the United States and Europe. Nosocomial infections are becoming more difficult to combat using conventional antibiotic therapies due to the increased incidence of multi-drug resistant bacteria. There is a growing need to find alternative therapeutic and preventative approaches.
- the gram-negative bacterium P. aeruginosa is a ubiquitous, opportunistic pathogen which displays a high level of antibiotic resistance and accounts for approximately 20% of nosocomial infections. P.
- aeruginosa poses a particular risk for the many hospitalized patients requiring mechanical ventilation, a number estimated at 800,000 annually in the United States alone (Wunsch et al, The epidemiology of mechanical ventilation use in the United States (2010) Crit. Care Med, 38: 1947-1953). In fact, patients with ventilator-associated pneumonia can have a mortality as high as 70-80% (Chastre and Fagon (2002) Ventilator-associated pneumonia Respir. Crit. Care Med.
- liver diseases hepatobiliary disorders
- P. aeruginosa hepatobiliary disorders
- liver diseases hepatobiliary disorders
- the severity of liver disease, the need for mechanical ventilation and high creatinine serum levels were found to be of highest prognostic significance; in fact, patients with all three factors had a 98% mortality.
- the present invention relates to the use of an OprF antigen in subjects with hepatobiliary disorders, a medical history of hepatobiliary disorders and/or an indication associated with hepatobiliary disorders and wherein the subject is, has been, or is at risk of being an intensive care unit (ICU) patient and/or a burn patient. Further, the invention relates to the use of an OprF antigen in subjects with no respiratory, thoracic or mediastinal disorders and wherein the subject is, has been, or is at risk of being an intensive care unit (ICU) patient and/or a burn patient. Further, the invention relates to the use of an OprF antigen in the manufacture of a medicament for the treatment and/or prevention of a P. aeruginosa infection in the subject. The subject to be treated may also be mechanically ventilated. The present invention also relates to the above uses, wherein the risk of mortality of the treated subject is reduced compared to a placebo-treated subject.
- the antigen for use in the methods and compositions of the invention may comprise an OprF antigen, derived from the outer membrane protein F (OprF) of P. aeruginosa.
- the OprF antigen of the invention may comprise or consist of the OprF polypeptide set forth as SEQ ID NO: 1 or an immunogenic variant or antigenic fragment thereof; may comprise or consist of the OprF/I fusion protein set forth as SEQ ID NO: 3 or an immunogenic variant or antigenic fragment thereof; or may comprise a mixture of the aforementioned polypeptides or immunogenic variants or antigenic fragments thereof.
- the OprF antigen for use in the methods and compositions of the invention need not be the polypeptide, immunogenic variant or antigenic fragment itself as set forth in this paragraph, but, alternatively or additionally, may be a molecule that can be used to or that causes expression of an OprF antigen as disclosed herein, e.g., when used as a medicament and/or in the treatment of a subject as described herein.
- Non-limiting examples of such a molecule include nucleic acids encoding OprF antigens or antigenic fragments or immunogenic variants of the invention.
- the antigen for use in the methods and compositions of the invention may be a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an immunogenic variant or antigenic fragment thereof; and/or a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an immunogenic variant or antigenic fragment thereof, optionally delivered with an adjuvant.
- the antigen of the invention is antigenic, i.e., capable of eliciting an immune response against the OprF antigen, and preferably, an immune response against Pseudomonas aeruginosa and/or Pseudomonas aeruginosa infection.
- an immune response against Pseudomonas aeruginosa and/or Pseudomonas aeruginosa infection preferably, an immune response against Pseudomonas aeruginosa and/or Pseudomonas aeruginosa infection.
- the OprF antigen may be an antigenic variant or fragment of a polypeptide comprising or consisting of SEQ ID NO: 1 and/or SEQ ID NO: 3, provided that the variant or fragment elicits or is capable of eliciting an immune response against an OprF antigen, an immune response against Pseudomonas aeruginosa and/or an immune response against a Pseudomonas aeruginosa infection; i.e., that the fragment or variant is antigenic or immunogenic.
- the antigenic variants of the OprF antigens as described herein may be polypeptides sharing at least 80% sequence identity with the OprF antigen, or may be a nucleic acid encoding a polypeptide sharing at least 80% sequence identity with the OprF antigen.
- the OprF antigen of the invention may be a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 1 or fragment thereof; may be a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 3 or fragment thereof; may be a nucleic acid molecule encoding a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 1 or fragment thereof; or may be a nucleic acid molecule encoding a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 3 or fragment thereof, provided that the OprF antigen elicits or is capable of eliciting an immune response against an OprF antigen, P. aeruginosa and/or a P. aeruginosa
- the methods described herein may be used to reduce mortality and/or to treat or prevent infection with P. aeruginosa in subjects who have a hepatobiliary disorder, a medical history of a hepatobiliary disorder or one or more markers of a hepatobiliary disorder and/or may be, or are at risk of becoming, intensive care unit patients and/or burn patients. Further, the methods may be used to reduce mortality and/or to treat or prevent infection with P.
- the methods and uses described herein are based, at least in part, on the surprising discovery that administration of an OprF antigen to ICU patients having 1) hepatobiliary disorders, a medical history of hepatobiliary disorders and/or markers of hepatobiliary disorders or 2) having no history of or existing respiratory, thoracic or mediastinal (RTM) disorder results in a sharp decrease in overall mortality relative to subjects that do not receive the treatment (i.e., placebo control group).
- the methods provided herein are useful for the treatment and/or prevention ⁇ . aeruginosa infection, which is particularly prevalent in ICU patients (especially
- the invention relates in particular to the use of OprF antigens as described herein for the treatment or prevention of infection with Pseudomonas aeruginosa, or a symptom thereof, wherein the subject to be treated is a hospitalized patient, especially an ICU patient, a patient with a hepatobiliary disorder, a history of a hepatobiliary disorder or with one or more markers of a hepatobiliary disorder, having no history of or existing RTM disorder, is at risk of becoming an ICU patient, a patient who is intubated or expected to be intubated, a burn patient and/or combinations thereof.
- the invention relates to the uses, methods and compounds disclosed herein wherein the subject to be treated is a patient with a hepatobiliary disorder, has a medical history of a hepatobiliary disorder and/or one or more markers of a hepatobiliary disorder, is an ICU patient and is mechanically ventilated.
- the compounds, uses and methods of the invention are effective to prevent or treat nosocomial bacterial infections, in particular P. aeruginosa infections, or symptoms or sequelae thereof.
- the treatment or prevention is achieved by inducing protective immunity and/or reducing the virulence of the infectious bacteria.
- the term "virulence” refers to the degree of pathogenicity of an organism as indicated by case fatality rates and/or the ability of the organism to invade the tissues of the host.
- the virulence of an organism is determined by its virulence factors; i.e., gene products produced in response to certain environmental signals, and its virulence state.
- Virulence can be understood not only in terms of proximate causes— those specific traits of the pathogen that help make the host ill— but also by ultimate causes— the evolutionary pressures that lead to virulent traits occurring in a pathogen strain ("Virulence”. Wikipedia: The Free
- the virulence state of, e.g., a bacterium can be determined by a complex interplay between host and bacterial factors.
- virulence can be defined as an infection-induced loss of host fitness.
- the virulence of a microorganism may be assessed by any number of parameters, or combinations thereof, as known in the art. Non-limiting examples of parameters used to assess
- microorganism virulence include adherence of the microorganism to cells or tissues of a subject (a host organism), colonization, invasion of cells or tissues, secretion of toxins resulting in death or damage of cells or tissues, formation of biofilms on cells or tissues, formation of biofilms on exogenous materials (i. e. , materials implanted temporarily or permanently in a subject), evasion or alteration of immune or other responses in the subject, resistance to antibiotics, scavenging of necessary nutrients or cofactors (e.g., iron), and/or motility.
- the virulence state of a microorganism such as P. aeruginosa is thus determined by a variety of factors, including genetic factors, induction of virulence factors and host responses, including specific and non-specific immune responses.
- a plausible mechanism by which administration of an OprF antigen of the invention results in reduced mortality is the impact of the host immune response to the OprF antigen on the virulence state of the bacterium, possibly mediated in part by antibodies interfering with OprF function. More generally, factors affecting bacterial virulence can be broadly classified as virulence inhibitors.
- Example of a host-produced virulence inhibitor in the context of the current invention may include OprF-specific antibodies (see Fig. 1 for a schematic representation of potential effects of host immunization on P. aeruginosa virulence). In this regard, Wu et al.
- aeruginosa infection exacerbates liver disease, in part by release of toxins such as pyocyanin, exotoxin A and endotoxin (LPS), leading to an increased risk of mortality in an already at- risk patient population (Chevaluppa, et al, International Journal of Infectious Diseases 14 (2010) e857-e867).
- toxins such as pyocyanin, exotoxin A and endotoxin (LPS)
- LPS endotoxin
- aeruginosa does not explain the observed P. aeruginosa infections in the vaccinated and non-vaccinated groups (i.e. there was not difference observed). This interpretation allows new prevention and treatment strategies of which some are explored and defined herein. E.g. with this hypothesis it can be explained why the hepatobiliary disorder subjects are surprisingly benefiting from the vaccination as herein proposed.
- Assessing effects of OprF antigen vaccination on the virulence of a P. aeruginosa can be carried out in vitro by, for example, quantifying the transcription of virulence factors or assessing other measures of virulence such as adherence or motility in the presence of host- produced factors.
- the host factors may be present, for example, in fluids of the immunized subject, such as, for example, blood, serum, sputum or urine.
- Assessing the interference of OprF function can be carried out in vitro by, for example, assessing the disruption of the OprF and OprF ligand (such as e.g. interferon gamma) interaction in an ELISA type assay.
- an "antigen” refers to a molecule or a fragment or portion of a molecule that is immunogenic; in other words, stimulates an immune response; e.g., promotes the production of specific antibodies or proliferation and/or activation of effector cells in the subject.
- the immune response is a humoral immune response; i.e., production of antibodies.
- the immune response is a cell-mediated immune response.
- the immune response is characterized by the production and/or release of immune regulating molecules; for example, cytokines or chemokines.
- the presence of an immune response can be determined by any number of techniques of common usage in the art; for example assessment of the serum levels of antigen-specific antibodies as measured by an increase in Geometric Mean Titer (GMT) and compared with sera from placebo-treated subjects or pre-immune serum from the same patient to determine seroconversion in response to vaccination. Briefly, GMT can be assessed by an in vitro test which shows the levels of antigen specific antibodies in the sera of patients.
- GTT Geometric Mean Titer
- the OprF antigen comprises or consists of a molecule that is able to stimulate the production of specific antibodies against OprF that are able to disrupt the binding of an OprF ligand to OprF (herein also generally referred to as "OprF antigen").
- the OprF antigen comprises or consists of the OprF polypeptide (SEQ ID NO: 1) or a fragment or a portion thereof.
- the OprF antigen is a portion of a fusion protein that includes a portion of at least one additional protein.
- the fusion protein is an OprF-OprI (OprF/I) fusion protein.
- OprF/I fusion proteins have been described, for example, in WO 2012/084272 and WO 2012/126879, herein incorporated by reference in their entirety.
- the OprF/I fusion proteins comprise a portion of the P.
- aeruginosa outer membrane protein F (OprF, SEQ ID NO: 1) fused via the carboxy terminal end to a portion of the P. aeruginosa outer membrane protein I (Oprl, SEQ ID NO: 2).
- the portion ⁇ . aeruginosa OprF comprises amino acids 190-342 of P. aeruginosa OprF and amino acids 21- 83 ⁇ . aeruginosa Oprl.
- the OprF/I protein comprises an N-terminal tag for purification purposes, such as a His-tag, especially His 6 , preceded N-terminally by an alanine residue.
- the N-terminal alanine residue is preceded N- terminally by a methionine residue.
- the N-terminal methionine residue is cleaved off during production of the OprF/I fusion protein.
- the OprF/I fusion protein comprises or consists of the polypeptide provided by SEQ ID NO: 3.
- the OprF/I fusion protein is an immunogenic variant of the OprF/I fusion protein provided by SEQ ID NO: 3.
- An immunogenic variant may comprise one or more amino acid variations in the sequence of the OprF/I fusion protein, provided that the variant maintains at least the same or not less than 80%, more preferably 85%, even more preferably 90%, even more preferably 95%, most preferred 98% of the immunogenic activity of the original molecule.
- Immunogenic activity is preferably assessed by determination of the GMT, as described above, in the sera of immunized subjects compared with pre-immune sera or sera from control (placebo) treated subjects.
- the amino acid variations may be mutations, such as, but not limited to, substitutions, insertions or deletions of residues, and may be made, for example, to enhance immunogenicity of the OprF antigen in the subject and/or the production of the OprF antigen (e.g. , by a recombinant cell).
- the amino acid variations are conservative amino acid residue substitutions.
- a "conservative amino acid substitution” is an amino acid substitution that does not alter the relative charge or size characteristics of the protein. Examples of conservative amino acid substitutions include the "conservatively modified variants" and “conservative sequence modifications" of polypeptides described in WO 2012/084272.
- the immunogenic variant retains the cysteine residues present at positions 18, 27, 33 and 47 of SEQ ID NO: 3.
- the OprF/I fusion protein comprises a variant polypeptide that is at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, or 99%) identical to the OprF/I polypeptide provided by SEQ ID NO: 3, wherein the variant polypeptide maintains at least the same or not less than 90%, not less than 85%, or not less than 80% of the immunogenic activity of SEQ ID NO: 3, wherein the immunogenic activity is assessed as outlined above.
- Two sequences are "substantially identical” if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (i. e., at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity) over a specified region or over the entire sequence, when compared and aligned for maximum correspondence over a comparison window, or designated region, as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection.
- the identity exists over a region that is at least about 30 nucleotides (or 10 amino acids) in length, or more preferably over a region that is 60 to 150 or 600 or more nucleotides (or 20, 50, 200 or more amino acids) in length.
- sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
- Methods of alignment of sequences for comparison are well known in the art; for example, the local homology algorithm of Smith and Waterman (Comparison of biosequences (1981) Adv. Appl. Math. 2:482 ⁇ 189), the homology alignment algorithm of Needleman and Wunsch (A general method applicable to the search for similarities in the amino acid sequence of two proteins (1970) J. Mol. Biol. 48:443-53), the search for similarity method of Pearson and Lipman (Improved tools for biological sequence comparison (1988) Proc. Natl. Acad. Sci. USA 85 :2444-8), also computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the
- any of the antigens described herein, such as the OprF/I fusion proteins, may be produced by any method known in the art.
- the antigen may be recombinantly produced in a eukaryotic or prokaryotic cell, such as a tissue culture cell, a bacterial cell, or a fungal or yeast cell; or may be synthesized by conventional means.
- a eukaryotic or prokaryotic cell such as a tissue culture cell, a bacterial cell, or a fungal or yeast cell; or may be synthesized by conventional means.
- the antigens are purified and processed as detailed in WO 2012/126879.
- nucleic acids encoding OprF antigens of the invention.
- the nucleic acids encode the OprF polypeptide (SEQ ID NO: 1) or an antigenic fragment thereof.
- nucleic acids encode the OprF/I fusion protein (SEQ ID NO: 3) or an antigenic fragment or immunogenic variant thereof.
- the protein or polypeptide encoded by the nucleic acid is produced by cells of the subject (e.g., using transcriptional and/or translational machinery of the cells of the subject). In such embodiments, the nucleic acid may further encode additional features known in the art for expression of the encoded antigen.
- the invention further encompasses OprF antigens as disclosed herein produced according to the following methods: recombinantly introducing an oligonucleotide encoding the OprF antigen into an expression vector, preferably pTRC99A, transforming a host cell, preferably E. coli, preferably E. coli strain XL1, and purifying and processing the resulting protein as detailed in WO 2012/126879, followed by storage at -80°C in PBS buffer.
- the processing of the recombinantly -produced OprF/I protein comprises a reduction step with dithiothreitol (dtt) and a controlled re-oxidation step in the presence of L-cystine.
- the thus processed OprF antigen is further purified before storage and/or formulation.
- An OprF antigen such as e.g. SEQ ID NO: 3 may be formulated e.g. as an injectable, such as for intramuscular or intravenous, preferably intramuscular administration, in a dose of 100 meg in a physiological salt solution (0.81% weight per volume) with or without aluminium hydroxide (400 meg).
- an OprF antigen such as e.g. SEQ ID NO: 3
- the subject is a mammalian subject, such as a human, non-human primate, rodent, rabbit, sheep, dog, cat, horse, llama or cow.
- the subject is a human subject, such as a patient.
- the invention relates to the compounds, uses and methods wherein the subject to be treated has a hepatobiliary disorder, a medical history of a hepatobiliary disorder or an indication or indications associated with a hepatobiliary disorder and/or is, has been, or is at risk of being, an intensive care unit patient.
- said subject is mechanically ventilated during at least part of a hospital or ICU stay.
- the subject to be treated is a burn patient.
- hepatobiliary disorders include any disorder that disrupts the normal function of the liver, gall bladder, bile duct and/or bile of the subject.
- hepatobiliary disorders include, without limitation, viral hepatitis (A, B or C or other), non- viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neon
- the subject with a hepatobiliary disorder, medical history of a hepatobiliary disorder and/or an indication or indications associated with a hepatobiliary disorder and/or is has been, or is at risk of being an intensive care unit patient and/or a burn patient additionally has no previous history of nervous system disorders.
- an indication associated with a hepatobiliary disorder may refer to the result of a clinical assessment, such as the level of a protein found in the serum, that is outside (above or below) the range of a result considered to be normal.
- clinical assessment such as the level of a protein found in the serum
- clinical relevant deviation or “clinically relevant levels” or “clinically relevant elevation” refers to a result of an assessment that is outside the range of a result considered to be normal and may indicate the subject has or is at risk of having a hepatobiliary disorder.
- the methods include testing a subject for a hepatobiliary disorder or indication associated with a hepatobiliary disorder.
- the subject may be tested for a hepatobiliary disorder or one or more indications associated with a hepatobiliary disorder prior to administration of the OprF antigen.
- Any assessment associated with or indicative of a hepatobiliary disorder or the dysfunction of the liver, gallbladder and/or bile duct may be compatible with the methods described herein. Examples of assessments include, without limitation, examining the medical history of the subject, performing a physical examination of the subject, measuring the level of one or more liver proteins in a serum sample from the subject, measuring hepatic excretory function, performing ultrasonography and/or computed tomography scanning of the liver, performing angiography and performing a liver biopsy.
- the presence of liver proteins in a serum sample indicates the subject has a hepatobiliary disorder.
- the presence of one or more liver proteins at a particular concentration indicates that the subject has a hepatobiliary disorder.
- the liver protein is serum glutamic pyruvic transaminase (SGPT; also referred to as alanine aminotransferase), serum glutamic oxaloacetic transaminase (SGOT; also referred to as aspartate aminotransferase), bilirubin, ⁇ - glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase and/or albumin.
- SGPT serum glutamic pyruvic transaminase
- SGOT serum glutamic oxaloacetic transaminase
- bilirubin ⁇ - glutamyltransferase
- alkaline phosphatase 5 '-nucleotidase and/or albumin.
- a Respiratory, Thoracic or Mediastinal (RTM) disorder is any disorder that affects the upper or lower respiratory tract and associated structures or any organs, systems, tissues or cells in the anatomical region of the thorax or mediastinum.
- RTM disorders may include, for example, any of the following diseases, symptoms or syndromes: acquired diaphragmatic eventration, pleural disorder, acute pulmonary edema, pleural effusion, acute respiratory distress syndrome, pleural fibrosis, acute respiratory failure, pleurisy, alveolitis allergic, pleuritic pain, apnea, pneumomediastinum, asphyxia, pneumonia aspiration, aspiration, pneumonitis, asthma, pneumothorax, asthma late onset, productive cough, pleural effusion, atelectasis, prolonged expiration, bronchial disorder, pulmonary alveolar hemorrhage, bronchial fistula, pulmonary arterial hypertension, bronchial hyperreactivity, pulmonary cavitation, bronchial obstruction, pulmonary congestion, bronchiectasis, pulmonary embolism, bronchitis chronic, pulmonary fibrosis, bronchopleural fistula, pulmonary hemorrhage, bronchos
- tracheomalacia lung disorder, upper airway resistance syndrome, lung infiltration, wheezing, lupus pneumonitis, mediastinal shift, obstructive airways disorder, oropharyngeal pain, oropharyngeal spasm, orthopnea, pharyngeal hemorrhage, pharyngeal edema, pickwickian syndrome, pleural calcification, laryngospasm, pulmonary microemboli, neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and organizing pneumonia.
- the methods include testing a subject for RTM disorders.
- RTM disorders can be assessed as deemed appropriate by the attending medical professional.
- the subject may be tested for an RTM disorder prior to administration of the OprF antigen. Any assessment associated with or indicative of an RTM may be compatible with the methods described herein. Examples of assessments include, without limitation, examining the medical history of the subject, performing a physical examination of the subject, X-ray imaging, chest auscultation, palpation, magnetic resonance imaging (MRI), electrocardiogram (ECG), echocardiogram, pulse oxiometry, computerized axial tomography (CAT) scan, lung function tests, e.g., spirometry, and blood testing.
- MRI magnetic resonance imaging
- ECG electrocardiogram
- ECG electrocardiogram
- oxiometry computerized axial tomography
- CAT computerized axial tomography
- ANP atrial natriuretic peptide
- BNP B-type natriuretic peptide
- the subject is an ICU patient and/or a burn patient, has been an ICU patient and/or a burn patient, or is at risk of being an ICU patient and/or burn patient.
- an ICU patient means that the subject has a medical condition requiring the support of an intensive care unit of a medical facility.
- the subject may have a severe and/or life-threatening medical condition and/or injuries. Whether a subject is an ICU patient and/or a burn patient will be evident to one of skill in the art.
- the subject has been an ICU patient, for example the subject was an ICU patient within the previous 1 day to 6 months prior to administration of any of the OprF antigens described herein.
- the subject was an ICU patient within at least the previous 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 25, 26, 27, 28, 29, 30 days, or at least 1, 2, 3, 4, 5, 6 months prior to administration of any of the OprF antigens described herein.
- the subject is or has been mechanically ventilated, for example using a positive pressure ventilator or a negative pressure ventilator.
- the subject is at risk of being an ICU patient.
- the subject has a disease, disorder, or injury that has the risk of progressing to a stage that requires the support of an intensive care unit of a medical facility.
- the subject may participate in activities that put the subject at risk of severe bodily injury that may require the support of an intensive care unit of a medical facility.
- Examples of activities that may put the subject at risk of severe injury include, without limitation, extreme sports, such as base jumping, bungee jumping, gliding, hang gliding, high wire, ski jumping, sky diving, sky surfing, sky flying, indoor climbing, adventure racing, aggressive inline skating, BMX, caving, extreme motocross, extreme skiing, freestyle skiing, land and ice yachting, mountain biking, mountain boarding, outdoor climbing, sandboarding, skateboarding, snowboarding, snowmobiling, speed biking, speed skiing, scootering, barefoot waterskiing, cliff diving, free-diving, jet skiing, open water swimming, powerboat racing, round the world yacht racing, scuba diving, snorkeling, speedsailing, surfing, wakeboarding, Whitewater kayaking, windsurfing.
- any of the OprF antigens described herein may be administered to the subject prior to participation in an activity that may put the subject at risk of being an intensive care unit patient.
- the subject may be, have been, or be at risk of being a burn patient.
- the subject has or is at risk of sustaining a burn that requires support from a specialized burn facility.
- the severity of burns is generally classified based on the depth of skin and tissues affected by the burn.
- the burn or burns may be first-degree, second- degree, third-degree, fourth-degree, or a combination thereof. Whether a subject is a burn patient will be evident to one of skill in the art.
- the subject has been a burn patient, for example the subject was a burn patient within the previous 1 day to 6 months prior to administration of any of the OprF antigens described herein.
- the subject was a burn patient within at least the previous 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 25, 26, 27, 28, 29, 30 days, or at least 1, 2, 3, 4, 5, 6 months prior to administration of any of the OprF antigens described herein.
- the subject is at risk of being a burn patient.
- the subject may participate in activities that put the subject at risk of sustaining a burn, for example a burn that may require the support of a specialized burn facility.
- the subject has an occupation that may put the subject at risk of sustaining a burn, for example, the subject is a firefighter, works in food preparation, or works with chemicals.
- any of the OprF antigens described herein may be administered to the subject prior to participation in an activity or occupation that may put the subject at risk of being a burn patient.
- the subject at risk of becoming an ICU patient or a burn patient is elderly, immunocompromised, catheterized, undergoing surgery, a cancer patient, an HIV- infected patient or a cystic fibrosis patient and, as such, particularly at high risk for P.
- these subjects may be administered any of the OprF antigens described herein at any time before admission or imminent admission into the ICU.
- the OprF antigen is administered to the subject once. In some embodiments, the OprF antigen is administered to the subject more than once. In some embodiments the OprF antigen is administered to the subject on at least 2, 3, 4, 5, 6, 7, 8, 9, 10 or more occasions. Any administration of the OprF antigen that is administered after the first administration may be referred to as a "booster".
- a subsequent second administration of the OprF antigen is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24,2 5, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or at least 60 days after the first administration of the OprF antigen (preferred for the second administration is 7 days after the first administration).
- a subsequent (subsequent to the second and any further administrations) administration of the OprF antigen is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24,2 5, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or at least 60 days after the prior administration of the OprF antigen.
- a subject is in need of one or more additional administrations of the OprF antigen will be evaluated according to one of ordinary skill in the art.
- the compounds, uses and methods described herein may result in a reduction of the mortality in a subject being administered the OprF antigen.
- “mortality” means death of a subject.
- “Survival” means non-death of a subject.
- the "overall survival” ("OS") of the subjects in a given group or “survival” can also be expressed as a percentage of survivors in the group at a given timepoint; for example, day 28 or day 90 survival.
- the survival is the inverse of the mortality. Any expression of mortality can also be expressed inversely as survival.
- the term "increased survival” or “increased overall survival” refers to a comparison of the survival of two different patient groups, especially to the difference in survival between treated and placebo patient groups, particularly at a given timepoint.
- An example of increased survival in the current invention is the survival in subjects who are administered an OprF antigen (e.g., a treated subject group) minus the survival in subjects who do not receive the treatment (i.e., a placebo control group) and in case where the survival in the treated group is higher than in the placebo group the difference is > 0% and thus the overall survival or survival is increased.
- an OprF antigen e.g., a treated subject group
- a placebo control group i.e., a placebo control group
- Increased survival of the subject if the ratio of the mortality of treated subjects (at e.g. d28) versus the mortality of placebo controlled subjects (at e.g. d28) times one hundred (xlOO) > 100; i.e.,
- the term "mortality” refers to a measure of the number of deaths in a target population under certain conditions (e.g., a subject with a past or ongoing hepatobiliary disorder that is an intensive care unit patient and that is administered the OprF/I fusion protein set forth as SEQ ID NO: 3) per unit time (e.g., 28 days from first administration of the OprF/I fusion protein set forth as SEQ ID NO: 3), expressed as a percentage.
- aspects of the invention describe e.g., the mortality; i.e., the number of deaths in the population of subjects within the period of the first treatment administration with e.g., the OprF/I fusion protein (day 0) until 28 days later (day 28; referred to herein as day 28 mortality) or until 90 days later (day 90; referred to herein as day 90 mortality) expressed as a percentage.
- the mortality i.e., the number of deaths in the population of subjects within the period of the first treatment administration with e.g., the OprF/I fusion protein (day 0) until 28 days later (day 28; referred to herein as day 28 mortality) or until 90 days later (day 90; referred to herein as day 90 mortality) expressed as a percentage.
- the term "reduced mortality” is defined as the difference in the mortality of a treated group in comparison with the mortality in a non-treated group (e.g., a placebo group), particularly at a given timepoint. More specifically, reduced mortality is when the difference in the mortality of placebo controlled patients compared with the mortality of drug treated patients (e.g., in the OprF/I treated patient group) is greater than 0%. For example: Mortality at day 28 (d28) is reduced if the mortality of placebo group at d28 minus mortality of treated group at d28 > 0%;
- Reduced mortality of the subject is true if the ratio of the mortality of treated subjects (at e.g. d28) versus the mortality of placebo controlled subjects (at e.g. d28) times one hundred (xlOO) ⁇ 100; i.e., (mortality of treated subjects [at e.g. d28]/mortality of placebo controlled subjects [at e.g. d28]) x 100 ⁇ 100
- the mortality of the subject being administered the OprF antigen is reduced relative to the mortality of a subject who is not administered the OprF antigen; i.e., the placebo control. In some embodiments, the mortality of the subject being administered the OprF antigen is reduced compared to a subject that did not receive treatment (i.e., received a placebo control) at day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85,
- the mortality of the subject being administered the OprF antigen is reduced compared to a subject that did not receive treatment at day 28 after administration. In some embodiments, the mortality of the subject being administered the OprF antigen is reduced compared to a subject that did not receive treatment at day 90 after administration.
- the mortality of the subject being administered the OprF antigen is reduced by at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 1 1%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59% or at least 60% compared to a subject that did not receive treatment.
- the reduced mortality of the subject being administered the OprF antigen is at least 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94 or at least 95 compared to a subject that did not receive treatment.
- the term "probability of dying” is defined as the probability of dying in a particular situation for a subject.
- the particular situation is a hepatobiliary subject being in, or shortly before being admitted to an ICU, benefiting from the treatment and preventative strategies disclosed and claimed herein and has a reduced probability of dying as shown herein.
- the reduced probability of dying of the subject correlates with the degree of reduction of observed mortality, i.e. the higher the reduced mortality, the lower the probability of dying of the subject. This observation is particularly surprising as mortality is reduced, but the overall Pseudomonas aeruginosa infection rate has not been reduced despite the vaccination against Pseudomonas aeruginosa (see also supra for a possible explanation of the mechanism of action).
- any of the methods or uses described herein may be for the treatment or prevention of a P. aeruginosa infection and/or a symptom thereof in a subject.
- the terms “treat” and “treating” shall mean the administration of an OprF antigen to a subject to cure, ameliorate, prevent, reduce, or delay the onset of the symptoms, complications, pathologies, sequelae or biochemical indicia of a disease or infection (such as e.g., prevent or delay infections in subjects), alleviating the symptoms or arresting or inhibiting further
- Treatment may be prophylactic, i.e. prevention or delay of onset of the disease, or prevention or reduction of manifestations of clinical or subclinical symptoms thereof or therapeutic, i.e. suppression or alleviation of symptoms after the manifestation of the disease.
- prevention of as used herein shall mean "protection from” in the sense that the severity of disease, in the case of infection, is reduced.
- An OprF antigen that is administered to a subject prophylactically e.g., including prior to infection but generally also including any administration where the infection is not severely affecting the health of the subject
- the subject has or is at risk of having hepatobiliary disorders, for example having one or more indicia of hepatobiliary disorders, as otherwise herein described.
- the subject has no history of or existing RTM disorders.
- the subject has a P. aeruginosa infection.
- P. aeruginosa may cause any of a variety of types of infections in a subject, for example, the infection may be of the respiratory tract (e.g., pneumonia), bloodstream (bacteremia), heart (endocarditis), central nervous system (e.g., meningitis, brain abscess), ear (e.g., otitis externa, otitis media), eye (e.g., bacterial keratitis, endophthalmitis), bones and joints (e.g., osteomyelitis),
- the respiratory tract e.g., pneumonia
- bloodstream bacteremia
- heart endocarditis
- central nervous system e.g., meningitis, brain abscess
- ear e.g., otitis externa, otitis media
- eye e.g., bacterial keratitis, endophthalmitis
- bones and joints e.g., osteo
- P. aeruginosa infections may be classified as acute or chronic infections.
- Acute P. aeruginosa infections are characterized as rapidly spreading, causing tissue damage and/or sepsis, and may be associated with a high mortality rate.
- Chronic P. aeruginosa infections are characterized as persisting for weeks, months or years.
- the subject has an acute P. aeruginosa infection.
- the subject has fulminant P.
- aeruginosa infection such as, for example, bacterial keratitis.
- "Fulminant” as applied to an infection and as used herein shall mean an aggressive acute disease condition; i.e., one which is characterized by rapid onset of a new infection or sudden and rapid worsening of an existing infection and which is especially severe and/or life-threatening.
- a fulminant infection can furthermore arise from chronic infections.
- the subject has a chronic P. aeruginosa infection.
- the P. aeruginosa infection may be the result of or caused by the subject having been mechanically ventilated.
- OprF antigens described herein may be administered to a subject with, prior to, or after administration of an adjuvant.
- An adjuvant is an agent that enhances a response in a subject, such as an immune response, to an antigen or other molecule.
- an adjuvant may stabilize an antigen or other molecule. Determining whether an OprF antigen is administered with an adjuvant depends on various factors (e.g., type and extent of response desired) and will be evident to one of skill in the art.
- adjuvants may include, without limitation, alum, IC31 ®, aluminum hydroxide, aluminum phosphate, calcium phosphate hydroxide, paraffin oil, killed bacteria, bacterial toxins, toxoids, subunits of bacteria, squalene, detergents, IL-1, IL-2, IL-12, TLR9 agonists and combinations such as Freund's complete adjuvant and Freund's incomplete adjuvant.
- the OprF antigens are administered with aluminum hydroxide. In some embodiments, the OprF antigens are administered without adjuvant.
- an "effective amount" of an OprF antigen is any amount that results in a desired response or outcome in a subject, such as those described herein, including but not limited to reduction of mortality or risk of mortality.
- the OprF antigen may be formulated for administration in a pharmaceutical composition.
- pharmaceutical composition as used herein means a product that results from the mixing or combining of one or more active ingredients and includes both fixed and non- fixed combinations of the one or more active ingredient(s).
- fixed combination means that the active ingredients, e.g., the OprF antigen and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage.
- non-fixed combination means that the active ingredients, e.g., the antigen and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides
- a pharmaceutical composition as used herein can also mean a vaccine or vaccine composition.
- compositions of the invention can be prepared in accordance with methods well-known and routinely practiced in the art (see e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Co. 20th ed. 2000; and Ingredients of Vaccines - Fact Sheet from the Centers for Disease Control and Prevention).
- compositions are preferably manufactured under GMP conditions.
- a pharmaceutically effective dose of the OprF antigen is employed in the pharmaceutical composition of the invention.
- the OprF antigen is formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Dosage regimens are adjusted to provide the optimal desired response (e.g., the therapeutic or prophylactic response).
- Dosage levels of the active ingredients in the pharmaceutical compositions of the present invention can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired pharmaceutical response for a particular subject, composition, and mode of administration, without being toxic to the subject.
- the selected dosage level depends upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the subject being treated and like factors.
- a physician, veterinarian or other trained practitioner can administer doses of the
- OprF antigen of the invention employed in the pharmaceutical composition at levels lower than that required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
- effective doses of the compositions of the present invention for the prophylactic and therapeutic treatment of groups of people as described herein vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is a human or an animal, other medications administered and whether treatment is prophylactic or therapeutic. Treatment dosages need to be titrated to optimize safety and efficacy.
- OprF/I agent such as a vaccine comprising an OprF/I agent
- OprF/I fusion protein the doses range between 10 and 1000 meg, 50 to 500 meg, 100 to 200 meg, especially 100 meg.
- An exemplary treatment regime entails systemic administration; e.g., twice or once for a vaccine. In a preferred embodiment, the vaccine is administered twice.
- systemic administration includes such routes of delivery as subcutaneous, intramuscular, intradermal, intranasal, intravaginal or intrarectal, preferably intramuscular.
- An exemplary treatment regime entails systemic administration at day 0 and day 7 for the vaccine consisting of 100-200 meg SEQ ID NO: 3 and approximately 0.9% weight per volume NaCl in water or PBS, and optionally containing alum, preferably without alum.
- the OprF/I fusion protein according to SEQ ID NO:3 as well as adjuvanted or non-adjuvanted formulations comprising the OprF/I fusion protein according to SEQ ID NO:
- Day 0 shall be defined as the day on which the subject receives the first vaccination which shall preferably be the day of admittance to the hospital or ICU or, alternatively, the day on which the condition of the patient necessitates mechanical ventilation.
- Figure 1 shows a schematic representation of potential effects of virulence inhibitors, which may include antibodies produced in response to IC43 as shown, on the virulence of Pseudomonas and potential outcomes of these effects during an infection.
- the image represents an OprF protein assembled in the outer membrane ⁇ . aeruginosa.
- Figure 2 shows the percent survival over time of all subjects in clinical trial IC43-201 following the administration of three different formulations of IC43 or placebo.
- the treatments included the following: 100 micrograms of the OprF/I fusion protein without adjuvant (IC43 100 meg w/o, 97 subjects), 100 micrograms of the OprF/I fusion protein with alum (IC43 100 meg, 107 subjects), 200 micrograms of the OprF/I fusion protein with alum (IC43 200 meg, 100 subjects) and alum only (Placebo, 98 subjects). Subjects were vaccinated on days 0 and 7 and survival was monitored. Censoring is indicated by the presence of crosses.
- Figure 3 shows the percent survival over time of the subset of subjects in the IC43- 201 trial who presented at the start of the trial with a previous medical history of
- Hepatobiliary disorders YES and IC43 MedHist Hepatobiliary disorders NO are compared with the placebo-treated groups (Placebo MedHist Hepatobiliary disorders YES and Placebo MedHist Hepatobiliary disorders NO).
- the p-value is with respect to the IC43-treated versus placebo groups of subjects with a medical history of hepatobiliary disorders and was calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
- Figure 4 shows the survival over time of the subset of patients in the IC43-201 trial with a previous medical history of hepatobiliary disease as in Figure 3, with the results of each of the IC43 formulations shown separately.
- the p-values are with respect to the IC43- treated versus placebo groups of subjects with a medical history of hepatobiliary disorders and were calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
- Figure 4A shows the survival plot of subjects in the IC43-201 trial following day 0 and day 7 administration of 100 micrograms of non-adjuvanted OprF/I fusion protein or placebo.
- Figure 4B shows the survival plot of subjects in the IC43-201 trial following administration of 100 micrograms of OprF/I fusion protein with adjuvant or placebo.
- Figure 4C shows the survival plot of subjects in the IC43-201 trial following administration of 200 micrograms of OprF/I fusion protein with adjuvant or placebo.
- Figure 5 shows the survival over time of the subset of patients in the IC43-201 trial with clinically-relevant elevations of serum levels of two liver enzymes: serum glutamic oxaloacetic transaminase (SGOT) and serum glutamic pyruvic transaminase (SGPT) following IC43 or placebo administration.
- SGOT serum glutamic oxaloacetic transaminase
- SGPT serum glutamic pyruvic transaminase
- Figure 5A shows the survival over time of the subset of subjects in the IC43-201 trial with clinically relevant (CR) SGOT levels at the start of the trial compared with those subjects with non-clinically relevant (NCR) SGOT levels following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo.
- the data from all three IC43 treatments (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) are combined and the overall survival of the two populations is shown (IC43 SGOT.V0 CR and IC43 SGOT.V0 NCR) and compared with the placebo-treated groups (Placebo SGOT.V0 CR and Placebo SGOT.V0 NCR).
- Subjects with a clinically relevant deviation of SGOT at base-line in the IC43 and placebo groups are shown (lighter solid lines and dashed lines, respectively) and patients without clinically relevant deviation of SGOT at base-line in the IC43 -treated and placebo cohorts are shown (darker solid lines and dashed lines, respectively). The number of patients in each group is indicated in the legend.
- Figure 5B shows the survival over time of the subset of subjects in the IC43-201 trial having CR SGPT levels at the start of the trial compared with those subjects with NCR SGPT levels following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo.
- the data from all three IC43 treatments (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) are combined and the survival over time of the two populations is shown (IC43 SGPT.VO CR and IC43 SGPT.VO NCR) and compared with the placebo-treated groups (Placebo SGPT.VO CR and Placebo SGPT.VO NCR).
- Subjects with clinically relevant deviation of SGPT at BL in the IC43 and placebo cohorts are shown (lighter solid lines and dashed lines, respectively) and patients without clinically relevant deviation of SGPT at BL in the IC43-treated and placebo cohorts are shown (darker solid lines and dashed lines, respectively).
- the number of patients in each group is indicated in the legend.
- Figure 5C shows the overlap between three patient subsets selected from all IC43- and placebo-treated patients in the IC43-201 trial: those with a previous medical history of hepatobiliary disorders (77), those with clinically-relevant SGOT serum levels (69) and those with clinically relevant SGPT serum levels (48).
- Figure 6A shows the survival over time of the subset of subjects in the IC43-201 trial with an abnormal hepatic physical examination as measured at baseline compared to subjects with a normal hepatic physical examination following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo.
- IC43 PhyExam Hepatic ABNORMAL and IC43 PhyExam Hepatic NORMAL Data from all three IC43 -treated groups (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) are combined and the survival over time of the two populations is shown (IC43 PhyExam Hepatic ABNORMAL and IC43 PhyExam Hepatic NORMAL) and compared with the placebo-treated groups (Placebo PhyExam Hepatic ABNORMAL and Placebo PhyExam Hepatic NORMAL). (The p-value is with respect to the IC43 -treated and placebo groups with an abnormal hepatic examination and was calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
- Figure 6B shows the overlap of two patient subsets from the IC43-201 trial: those with an abnormal hepatic physical examination (88 in total) and those with a previous medical history of hepatobiliary disorders (77 in total).
- Figure 8A shows the survival over time of the subset of subjects in the IC43-201 trial with no medical history of respiratory, thoracic or mediastinal disorders at baseline compared to subjects with a medical history of respiratory, thoracic or mediastinal disorders following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo.
- IC43 OprF/I fusion protein
- Figure 8B shows the overlap of two patient subsets in the IC43-201 trial: those with no medical history of respiratory, thoracic or mediastinal disorders (153 in total) and those with a previous medical history of hepatobiliary disorders (77 in total).
- the amino acid sequence of OprF is provided by SEQ ID NO: 1 :
- the amino acid sequence of Oprl is provided by SEQ ID NO: 2:
- the amino acid sequence of the OprF/I fusion protein (IC43) is provided by SEQ ID NO: 3
- Clinical trial IC43-201 Clinical trial IC43-202
- aeruginosa infection in (ICU) Overall survival in all patients and in patients surviving Day 14 patients receiving IC43 vaccination versus Day 28 all cause mortality in patients with documented medical placebo during ICU stay history of a hepatobiliary disorder receiving IC43 or placebo
- aeruginosa such as bacteremia (determined as positive blood culture) or P. aeruginosa urinary tract infection in patients receiving IC43 or placebo within 28 and 56, 90 and 180 days after first vaccination
- Hepatobiliary disorders Hepatic steatosis
- Hepatobiliary disorders Hepatitis alcoholic
- Hepatobiliary disorders Hepatic ischaemia
- SOC System organ class
- SOC System organ class
- a Phase II placebo-controlled clinical trial (IC43-201) was performed including 399 male and female patients randomized into four cohorts at 33 different medical centers. The patients in the study had been admitted to an ICU with a need for mechanical ventilation for more than 48 hours and were between 18 and 80 years of age (mean 56.1 years). The subjects were determined by a clinician to have a high probability of survival for at least 48 hours.
- SOFA Sequential Organ Failure Assessment
- the P. aeruginosa OprF/I vaccine comprised a fusion protein of the
- Pseudomonas outer membrane porin proteins OprF and Oprl It was expressed as a 224 aa hybrid protein containing an N-terminal His 6 -tag which is preceded by an alanine and a methionine residue. The N-terminal methionine is cleaved off after expression in E.coli.
- the primary structure of the expressed fusion protein (excluding the N-terminal methionine) is shown in SEQ ID NO: 3.
- the OprF/I fusion protein was recombinantly produced in E. coli. Briefly, the OprF/I protein coding sequence was cloned into the multiple cloning site of the expression vector pTRC99A and transformed into E. coli strain XL1. The resulting protein was purified and processed as detailed in WO 2012/126879. The purified OprF/I protein was stored at -80°C in PBS buffer.
- the IC43 Drug Product with aluminium hydroxide consisted of the recombinant fusion-protein OprF/I adsorbed to the adjuvant aluminium hydroxide (Alhydrogel 2%).
- the drug substance comprising Ala-(His)6- OprFi 9 o-342-OprI 2 i-83 protein (OprF/I; SEQ ID NO: 3) in PBS buffer, was thawed overnight at 2-8°C and diluted 1 : 10 with physiological NaCl 0.9% solution to yield a final protein concentration of 100 mcg/mL, followed by 0.2 ⁇ sterile filtration.
- Sterile Alhydrogel 2% was added to a final concentration of 400 mcg/mL.
- the IC43 Drug Product without aluminium hydroxide was diluted 1 : 10 with NaCl 0.9% solution to yield a final protein concentration of 100 mcg/mL, followed by 0.2 ⁇ sterile filtration.
- the placebo was mixed with NaCl 0.9% solution.
- the placebo consisted of PBS diluted tenfold with 0.9% Saline, with 400 mcg/ml Al(OH)3 added.
- the 1 mL dose aliquots of 1.2 mL (extractable volume 1 mL) were aseptically filled into sterile pyrogen-free glass vials. All formulations were stored at 2-8°C before use.
- the patients in the groups receiving 100 meg IC43 were administered 1 mL of the formulations (with or without alum), whereas those patients receiving 200 meg IC43 were administered 2 mL of the formulation (with alum).
- Patients in the placebo group received 1 mL of placebo formulation.
- Randomized, placebo-controlled, partially -blinded phase 2 pilot study design 400 male or female patients admitted to an ICU with need for mechanical ventilation for more than 48 hours, aged between 18 and 80 years were vaccinated intramuscularly on days zero and seven in four treatment groups receiving 100 or 200 meg alum-adjuvanted OprF/I Vaccine (SEQ ID NO: 3), 100 meg non-adjuvanted OprF/I Vaccine (SEQ ID NO: 3) or alum as placebo control (see Table 1, below). Study duration per patient was estimated to be 90 days and overall study duration was estimated to be 12 to 18 months. Primary and secondary endpoints measured were as set forth in Table A2.
- Table 1 The number of subjects in the IC43-201 trial and the treatments received.
- the patients were intramuscularly administered a dose of 100 meg non-adjuvanted OprF/I vaccine (SEQ ID NO: 3; 97 patients); 100 meg alum-adjuvanted OprF/I vaccine (SEQ ID NO: 3; 104 patients); 200 meg alum-adjuvanted OprF/I vaccine (SEQ ID NO: 3; 100 patients); or alum only as placebo control (98 patients) (see Table 1).
- 100 meg non-adjuvanted OprF/I vaccine SEQ ID NO: 3; 97 patients
- 100 meg alum-adjuvanted OprF/I vaccine SEQ ID NO: 3; 104 patients
- 200 meg alum-adjuvanted OprF/I vaccine SEQ ID NO: 3; 100 patients
- alum only as placebo control 98 patients
- the study duration per patient was estimated to be 90 days and the overall study duration was estimated to be 12 to 18 months.
- Patients were evaluated for baseline parameters day 0 (visit 0, V0; see Table 3A), and for during-study parameters on days 7 (VI), 14 (V2), 28 (V3), 42 (V4), 56 (V5), 70 (V6), 90 (V7), and at the time of their ICU discharge (Va) (see Table 3B). Survival at day 28 was selected as a primary endpoint, and survival at day 90 was selected as a secondary endpoint.
- Subjects were evaluated for Sequential Organ Failure Assessment (SOFA) scores at each of the time points (data not shown).
- SOFA Sequential Organ Failure Assessment
- Table 3A Baseline parameters Medical history 25 Hepatobiliary, respiratory, skin disorders
- ICU 4 Reason total number of days in ICU
- Ventilation 3 Reason total number of days with ventilation
- the survival and mortality data for all three groups that received IC43 were combined and compared with placebo-treated subjects.
- a hierarchical clustering of subjects was performed to determine whether there were particular parameters, such as parameters related to medical history, laboratory measures, physical examination, or specific immune profiles, or subgroups within the subject population that were associated with enhanced or diminished survival relative to the overall population of subjects or to the population of subjects without the given parameter.
- the medical history of each subject participating in the IC43-201 trial was recorded at the start of the trial.
- any underlying disease, related symptoms as well as other pre-existing conditions prior to first vaccination were considered medical history.
- the medical history of each subject was reviewed by sites' study staff members at the time of the first study visit and, in this ICU trial, usually based on information provided by the medical record of the patient. Occasionally, site staff became aware of (and reported) single medical history events during the course of the study.
- Reported medical history was coded by the Contract Research Organization (CRO) Assign using the preferred term (PT) according to the Medical Dictionary for Regulatory Activities (MedDRA).
- CRO Contract Research Organization
- PT preferred term
- MedDRA Medical Dictionary for Regulatory Activities
- Table 5 Survival and mortality of subsets of patients in the IC43-201 trial with or without a previous medical history of hepatobiliary disorders after receiving the OprF/I vaccine or placebo. Data for all three IC43 treatment groups were combined and compared with placebo-treated subjects in each of the two groups (with and without a history of
- FIG. 5C shows the overlap between participating patient groups with clinically relevant (CR) elevations in serum SGOT and SGPT levels and those with a medical history of hepatobiliary disorders.
- 24 patients were characterized by all three parameters, whereas 91 patients were characterized by at least one of the parameters associated with disorders in the hepatobiliary system.
- Serum levels of SGOT and SGPT were assessed at each study site's local lab. In cases where the test results were not within the normal range specified by the respective lab, the investigator further assessed the result as being either CR or NCR. As an example, abnormal results requiring medical treatment or further diagnostics should be considered CR and thus be documented as an adverse event.
- normal SGPT and SGOT levels are approximately 10 to 50 units per liter in males and approximately 10 to 35 units per liter serum in female; however, normal ranges differ depending on the protocols and techniques used. During the study, each laboratory routinely provided the normal reference ranges and included them in the report.
- Figure 6A shows the overlap between participating patient groups with an abnormal hepatic exam and those with a medical history of hepatobiliary disorders. The total number of patients with an abnormal hepatic
- a hepatic physical examination in general can include palpation of the liver, estimation of the size of the liver and assessment of the patient for other visible and/or palpable signs and/or symptoms of liver disease.
- Shown in Figure 7 are the categories of hepatobiliary disorders identified in the patients taking part in the clinical trial, as well as the number of patients that were affected by the respective disorder.
- Example 1 Here will go the same types of data as outlined in Example 1; such as:
- Table x The number of subjects in the IC43-202 trial and the treatments received.
- Table x Investigator-confirmed P. aeruginosa infections arising during the course of the IC43-202 trial shown according to those that were identified in the first week, the second week or at any timepoint between the third week until the end of 90 days. Shown are numbers of patients developing invasive infection over the course of the study (including cases of bacteremia and/or pneumonia) as well as overall infection rates over the course of the study (DS). Overall infection numbers include confirmed bacteremia, pneumonia,
- a method of reducing the probability of dying in a subject comprising administering an effective amount of an OprF antigen to the subject; and/or a method for reducing the mortality of a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder, comprising administering an effective amount of an OprF antigen to the subject.
- the OprF antigen for use according to preferred aspect Al wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
- A3 The method according to preferred aspect Al or A2, wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject.
- A4. The method according to preferred aspects Al to A3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient.
- A5. The method according to preferred aspects Al to A4, wherein the subject has an existing Pseudomonas aeruginosa infection or is at risk of acquiring a Pseudomonas aeruginosa infection.
- A6 The method according to preferred aspect A5, wherein the existing or acquired Pseudomonas aeruginosa infection is a fulminant Pseudomonas aeruginosa infection.
- A7 The method according to any one of preferred aspects Al to A6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
- liver protein is alanine aminotransferase (SGPT), aspartate aminotransferase (SGOT), bilirubin, ⁇ - glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase, and/or albumin.
- SGPT alanine aminotransferase
- SGOT aspartate aminotransferase
- bilirubin ⁇ - glutamyltransferase
- alkaline phosphatase alkaline phosphatase
- 5 '-nucleotidase 5 '-nucleotidase
- the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO : 3.
- hepatobiliary disorder is viral hepatitis (A, B or C or other), non-viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, porphy
- B 1 The use of an OprF antigen in the manufacture of a pharmaceutical composition for reducing the probability of dying in a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder, comprising administering an effective amount of an OprF antigen to the subject; and/or the use of an OprF antigen in the manufacture of a pharmaceutical composition for reducing the mortality of a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder.
- B4 The use according to any of preferred aspects Bl to B3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient.
- B5. The use according to any of preferred aspects Bl to B4, wherein the subject has an existing bacterial infection or is at risk of acquiring a bacterial infection.
- liver protein is alanine aminotransferase (SGPT), aspartate aminotransferase (SGOT), bilirubin, ⁇ - glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase, and/or albumin.
- SGPT alanine aminotransferase
- SGOT aspartate aminotransferase
- bilirubin ⁇ - glutamyltransferase
- alkaline phosphatase alkaline phosphatase
- 5 '-nucleotidase 5 '-nucleotidase
- albumin albumin
- the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
- B 13 The use according to preferred aspects Bl 1 or B 12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200 ⁇ g/mL OprF antigen.
- B14 The use according to any one of preferred aspects B l to B 13, further comprising administering an adjuvant, preferably alum.
- aeruginosa infection is an acute P. aeruginosa infection, wherein the P. aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P. aeruginosa infection.
- hepatobiliary disorder is viral hepatitis (A, B or C or other), non-viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, porphy
- a pharmaceutical composition comprising an OprF antigen for reducing the probability of dying in a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder, comprising administering an effective amount of an OprF antigen to the subject; and/or the pharmaceutical composition comprising an OprF antigen for reducing the mortality of a subject, wherein the subject has an existing hepatobiliary disorder, a history of a
- hepatobiliary disorder or an indication associated with a hepatobiliary disorder.
- the pharmaceutical composition according to preferred aspect CI wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
- the pharmaceutical composition according to preferred aspect C2 wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject.
- composition according to any of preferred aspects CI to C4, wherein the subject has an existing bacterial infection or is at risk of acquiring a bacterial infection.
- C6 The pharmaceutical composition according to preferred aspect C5, wherein the existing or acquired bacterial infection is a Pseudomonas aeruginosa infection.
- C7 The pharmaceutical composition according to any one of preferred aspects CI to C6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
- composition according to any one of preferred aspects CI to C8, wherein said subject has been evaluated for said existing hepatobiliary disorder, history of hepatobiliary disorder or indication associated with hepatobiliary disorder, especially wherein said evaluation comprises at least one of the following
- liver protein is alanine aminotransferase (SGPT), aspartate aminotransferase (SGOT), bilirubin, ⁇ - glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase, and/or albumin.
- SGPT alanine aminotransferase
- SGOT aspartate aminotransferase
- bilirubin ⁇ - glutamyltransferase
- alkaline phosphatase 5 '-nucleotidase
- albumin albumin
- the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
- composition according to preferred aspect CI 1 wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
- composition according to preferred aspects CI 1 or C12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200 ⁇ g/mL OprF antigen.
- composition according to any one of preferred aspects CI to C13, further comprising administering an adjuvant, preferably alum.
- composition according to any one of preferred aspects C6 to CI 4, wherein the P. aeruginosa infection is an acute P. aeruginosa infection, wherein the P.
- aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P. aeruginosa infection.
- hepatobiliary disorder is viral hepatitis (A, B or C or other), non-viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis
- Dl A method of reducing the probability of dying in a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders, comprising administering an effective amount of an OprF antigen to the subject; and/or a method for reducing the mortality of a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders, comprising administering an effective amount of an OprF antigen to the subject.
- the OprF antigen for use according to preferred aspect Dl wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
- D5 The method according to preferred aspects Dl to D4, wherein the subject has an existing Pseudomonas aeruginosa infection or is at risk of acquiring a Pseudomonas aeruginosa infection.
- D6 The method according to preferred aspect D5, wherein the existing or acquired Pseudomonas aeruginosa infection is a fulminant Pseudomonas aeruginosa infection.
- D7 The method according to any one of preferred aspects Dl to D6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
- D8 The method according to preferred aspect D7, wherein the second administration of the OprF antigen is seven days after the first.
- D9 The method according to any one of preferred aspects Dl to D8, wherein said subject has been evaluated for said existing or medical history of respiratory, thoracic or mediastinal disorders, especially wherein said evaluation comprises at least one of the following
- ECG -electrocardiogram
- blood testing includes assessing arterial blood gas concentration and/or levels of indicator hormones, e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
- indicator hormones e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
- Dl 1 The method according to any one of preferred aspects Dl to D10, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3. D12.
- D12 The method according to preferred aspect D l 1, wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
- D13 The method according to preferred aspects Dl 1 or D 12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200 ⁇ g/mL OprF antigen.
- D14 The method according to any one of preferred aspects Dl to D13, further comprising administering an adjuvant, preferably alum.
- D15 The adjuvant, preferably alum.
- the respiratory, thoracic or mediastinal disorder is selected from the following: acquired diaphragmatic eventration, pleural disorder, acute pulmonary edema, pleural effusion, acute respiratory distress syndrome, pleural fibrosis, acute respiratory failure, pleurisy, alveolitis allergic, pleuritic pain, apnea, pneumomediastinum, asphyxia, pneumonia aspiration, aspiration, pneumonitis, asthma, pneumothorax, asthma late onset, productive cough, pleural effusion, atelectasis, prolonged expiration, bronchial disorder, pulmonary alveolar hemorrhage, bronchial fistula, pulmonary arterial hypertension, bronchial hyperreactivity, pulmonary cavitation, bronchial obstruction, pulmonary congestion, bronchiectasis, pulmonary embolism, bronchitis chronic, pulmonary fibrosis, bronchopleural fis
- tracheomalacia lung disorder, upper airway resistance syndrome, lung infiltration, wheezing, lupus pneumonitis, mediastinal shift, obstructive airways disorder, oropharyngeal pain, oropharyngeal spasm, orthopnea, pharyngeal hemorrhage, pharyngeal edema, pickwickian syndrome, pleural calcification, laryngospasm, pulmonary microemboli, neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and/or organizing pneumonia.
- pulmonary microemboli neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and/or organizing pneumonia.
- an OprF antigen in the manufacture of a pharmaceutical composition for reducing the probability of dying in a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders comprising administering an effective amount of an OprF antigen to the subject; and/or the use of an OprF antigen in the manufacture of a pharmaceutical composition for reducing the mortality of a subject, wherein the subject has an no existing or medical history of respiratory, thoracic or mediastinal disorders.
- the OprF antigen for use according to preferred aspect El wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
- E6 The use according to preferred aspect E5, wherein the existing or acquired bacterial infection is a Pseudomonas aeruginosa infection.
- E7 The use according to any one of preferred aspects El to E6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
- E8 The use according to preferred aspect E7, wherein the second administration of the OprF antigen is seven days after the first.
- E9 The use according to any one of preferred aspects El to E8, wherein said subject has been evaluated for an existing or medical history of respiratory, thoracic or mediastinal disorders, especially wherein said evaluation comprises at least one of the following
- ECG -electrocardiogram
- blood testing includes assessing arterial blood gas concentration and/or levels of indicator hormones, e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
- indicator hormones e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
- the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
- E14 The use according to any one of preferred aspects El to E13, further comprising administering an adjuvant, preferably alum.
- E15 The use according to any one of preferred aspects E6 to E14, wherein the P.
- aeruginosa infection is an acute P. aeruginosa infection, wherein the P. aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P. aeruginosa infection.
- the respiratory, thoracic or mediastinal disorder is selected from the following: acquired diaphragmatic eventration, pleural disorder, acute pulmonary edema, pleural effusion, acute respiratory distress syndrome, pleural fibrosis, acute respiratory failure, pleurisy, alveolitis allergic, pleuritic pain, apnea, pneumomediastinum, asphyxia, pneumonia aspiration, aspiration, pneumonitis, asthma, pneumothorax, asthma late onset, productive cough, pleural effusion, atelectasis, prolonged expiration, bronchial disorder, pulmonary alveolar hemorrhage, bronchial fistula, pulmonary arterial hypertension, bronchial hyperreactivity, pulmonary cavitation, bronchial obstruction, pulmonary congestion, bronchiectasis, pulmonary embolism, bronchitis chronic, pulmonary fibrosis, bronchopleural
- tracheomalacia lung disorder, upper airway resistance syndrome, lung infiltration, wheezing, lupus pneumonitis, mediastinal shift, obstructive airways disorder, oropharyngeal pain, oropharyngeal spasm, orthopnea, pharyngeal hemorrhage, pharyngeal edema, pickwickian syndrome, pleural calcification, laryngospasm, pulmonary microemboli, neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and/or organizing pneumonia.
- a pharmaceutical composition comprising an OprF antigen for reducing the probability of dying in a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders, comprising administering an effective amount of an OprF antigen to the subject; and/or the pharmaceutical composition comprising an OprF antigen for reducing the mortality of a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders.
- composition according to preferred aspect F2 wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject.
- F5. The pharmaceutical composition according to any of preferred aspects F l to F4, wherein the subject has an existing bacterial infection or is at risk of acquiring a bacterial infection.
- composition according to preferred aspect F5 wherein the existing or acquired bacterial infection is a Pseudomonas aeruginosa infection.
- F7 The pharmaceutical composition according to any one of preferred aspects Fl to F6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
- F8 The pharmaceutical composition according to preferred aspect F7, wherein the second administration of the OprF antigen is seven days after the first.
- ECG -electrocardiogram
- indicator hormones e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
- the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
- the pharmaceutical composition according to preferred aspect Fl 1, wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
Landscapes
- Chemical & Material Sciences (AREA)
- Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Medicinal Chemistry (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Immunology (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Genetics & Genomics (AREA)
- Microbiology (AREA)
- Gastroenterology & Hepatology (AREA)
- Mycology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
Abstract
Described herein are methods for administering Opr F antigens to a subject, wherein the subject has a hepatobiliary disorder, a medical history of a hepatobiliary disorder or one or more indicators of a hepatobiliary disorder.
Description
PSEUDOMONAS VACCINE
FIELD OF THE INVENTION
The disclosure relates to methods for the reduction of mortality and/or the treatment and/or prevention of Pseudomonas aeruginosa infection in subjects with hepatobiliary disorders and/or in patients with no respiratory, thoracic or mediastinal disorders.
BACKGROUND OF THE INVENTION Nosocomial infections, or hospital-acquired infections, are a significant cause of morbidity and mortality in both the United States and Europe. Nosocomial infections are becoming more difficult to combat using conventional antibiotic therapies due to the increased incidence of multi-drug resistant bacteria. There is a growing need to find alternative therapeutic and preventative approaches. The gram-negative bacterium P. aeruginosa is a ubiquitous, opportunistic pathogen which displays a high level of antibiotic resistance and accounts for approximately 20% of nosocomial infections. P. aeruginosa poses a particular risk for the many hospitalized patients requiring mechanical ventilation, a number estimated at 800,000 annually in the United States alone (Wunsch et al, The epidemiology of mechanical ventilation use in the United States (2010) Crit. Care Med, 38: 1947-1953). In fact, patients with ventilator-associated pneumonia can have a mortality as high as 70-80% (Chastre and Fagon (2002) Ventilator-associated pneumonia Respir. Crit. Care Med.
165:867-903).
Among patient populations susceptible to hospital-acquired infections such as P. aeruginosa, patients with hepatobiliary disorders (liver diseases) are particularly vulnerable to mortality when admitted to the ICU. A study following 100 patients with serious liver disease admitted to the ICU reported an overall mortality of 64% (Shellman, et al. Prognosis of patients with cirrhosis and chronic liver disease admitted to the medical intensive care unit (1988) Crit. Care Med. 16:671-678). In this study, the severity of liver disease, the need for mechanical ventilation and high creatinine serum levels were found to be of highest prognostic significance; in fact, patients with all three factors had a 98% mortality.
Additionally, patients with cirrhosis, a common form of liver disease, have particularly high mortality when admitted to the ICU— studies over several years have reported from 37% to 98% mortality in this group (Foreman, et al. Cirrhosis as a risk factor for sepsis and death (2003) Chest 124: 1016-1020).
In summary, there exists a clear medical need for improved preventative measures against nosocomial infections, particularly P. aeruginosa infections, in vulnerable patient populations such as patients with hepatobiliary disorders. SUMMARY
The present invention relates to the use of an OprF antigen in subjects with hepatobiliary disorders, a medical history of hepatobiliary disorders and/or an indication associated with hepatobiliary disorders and wherein the subject is, has been, or is at risk of being an intensive care unit (ICU) patient and/or a burn patient. Further, the invention relates to the use of an OprF antigen in subjects with no respiratory, thoracic or mediastinal disorders and wherein the subject is, has been, or is at risk of being an intensive care unit (ICU) patient and/or a burn patient. Further, the invention relates to the use of an OprF antigen in the manufacture of a medicament for the treatment and/or prevention of a P. aeruginosa infection in the subject. The subject to be treated may also be mechanically ventilated. The present invention also relates to the above uses, wherein the risk of mortality of the treated subject is reduced compared to a placebo-treated subject.
The antigen for use in the methods and compositions of the invention may comprise an OprF antigen, derived from the outer membrane protein F (OprF) of P. aeruginosa. The OprF antigen of the invention may comprise or consist of the OprF polypeptide set forth as SEQ ID NO: 1 or an immunogenic variant or antigenic fragment thereof; may comprise or consist of the OprF/I fusion protein set forth as SEQ ID NO: 3 or an immunogenic variant or antigenic fragment thereof; or may comprise a mixture of the aforementioned polypeptides or immunogenic variants or antigenic fragments thereof.
The OprF antigen for use in the methods and compositions of the invention need not be the polypeptide, immunogenic variant or antigenic fragment itself as set forth in this paragraph, but, alternatively or additionally, may be a molecule that can be used to or that causes expression of an OprF antigen as disclosed herein, e.g., when used as a medicament and/or in the treatment of a subject as described herein. Non-limiting examples of such a molecule include nucleic acids encoding OprF antigens or antigenic fragments or immunogenic variants of the invention. Therefore, alternatively or additionally, the antigen for use in the methods and compositions of the invention may be a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an immunogenic variant or antigenic fragment thereof; and/or a nucleic acid encoding the OprF/I fusion protein set forth as SEQ
ID NO: 3 or an immunogenic variant or antigenic fragment thereof, optionally delivered with an adjuvant.
As described herein, the antigen of the invention is antigenic, i.e., capable of eliciting an immune response against the OprF antigen, and preferably, an immune response against Pseudomonas aeruginosa and/or Pseudomonas aeruginosa infection. As used herein,
"immunogenic" means antigenic. The capability of an antigen of the invention to elicit an immune response against a desired target can be determined by any in vitro or in vivo method known in the art and/or described herein. The OprF antigen may be an antigenic variant or fragment of a polypeptide comprising or consisting of SEQ ID NO: 1 and/or SEQ ID NO: 3, provided that the variant or fragment elicits or is capable of eliciting an immune response against an OprF antigen, an immune response against Pseudomonas aeruginosa and/or an immune response against a Pseudomonas aeruginosa infection; i.e., that the fragment or variant is antigenic or immunogenic.
The antigenic variants of the OprF antigens as described herein may be polypeptides sharing at least 80% sequence identity with the OprF antigen, or may be a nucleic acid encoding a polypeptide sharing at least 80% sequence identity with the OprF antigen. For example, the OprF antigen of the invention may be a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 1 or fragment thereof; may be a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 3 or fragment thereof; may be a nucleic acid molecule encoding a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 1 or fragment thereof; or may be a nucleic acid molecule encoding a polypeptide comprising or consisting of an amino acid sequence having at least 80% sequence identity with the amino acid sequence set forth as SEQ ID NO: 3 or fragment thereof, provided that the OprF antigen elicits or is capable of eliciting an immune response against an OprF antigen, P. aeruginosa and/or a P. aeruginosa infection. DETAILED DESCRIPTION OF THE INVENTION
Disclosed herein are methods of administering an OprF antigen to a subject, and related uses and compositions. The methods described herein may be used to reduce mortality and/or to treat or prevent infection with P. aeruginosa in subjects who have a hepatobiliary disorder, a
medical history of a hepatobiliary disorder or one or more markers of a hepatobiliary disorder and/or may be, or are at risk of becoming, intensive care unit patients and/or burn patients. Further, the methods may be used to reduce mortality and/or to treat or prevent infection with P. aeruginosa in subjects who have no history of or existing respiratory, thoracic or mediastinal disorder and/or may be, or are at risk of becoming, intensive care unit patients and/or burn patients. The methods and uses described herein are based, at least in part, on the surprising discovery that administration of an OprF antigen to ICU patients having 1) hepatobiliary disorders, a medical history of hepatobiliary disorders and/or markers of hepatobiliary disorders or 2) having no history of or existing respiratory, thoracic or mediastinal (RTM) disorder results in a sharp decrease in overall mortality relative to subjects that do not receive the treatment (i.e., placebo control group).
The methods provided herein are useful for the treatment and/or prevention ΐΡ. aeruginosa infection, which is particularly prevalent in ICU patients (especially
mechanically-ventilated) and burn patient populations. As used herein, "mechanical ventilation" shall mean any method or technique of assisting the breathing of a patient, particularly, but not limited to, methods or techniques requiring intubation of the patient. Accordingly, the invention relates in particular to the use of OprF antigens as described herein for the treatment or prevention of infection with Pseudomonas aeruginosa, or a symptom thereof, wherein the subject to be treated is a hospitalized patient, especially an ICU patient, a patient with a hepatobiliary disorder, a history of a hepatobiliary disorder or with one or more markers of a hepatobiliary disorder, having no history of or existing RTM disorder, is at risk of becoming an ICU patient, a patient who is intubated or expected to be intubated, a burn patient and/or combinations thereof. Preferably the invention relates to the uses, methods and compounds disclosed herein wherein the subject to be treated is a patient with a hepatobiliary disorder, has a medical history of a hepatobiliary disorder and/or one or more markers of a hepatobiliary disorder, is an ICU patient and is mechanically ventilated.
The compounds, uses and methods of the invention are effective to prevent or treat nosocomial bacterial infections, in particular P. aeruginosa infections, or symptoms or sequelae thereof. Although not wishing to be bound by any particular theory, it is believed that the treatment or prevention is achieved by inducing protective immunity and/or reducing the virulence of the infectious bacteria. As used herein, the term "virulence" refers to the degree of pathogenicity of an organism as indicated by case fatality rates and/or the ability of the organism to invade the tissues of the host. The virulence of an organism is determined by its virulence factors; i.e., gene products produced in response to certain environmental
signals, and its virulence state. An organism producing higher levels of virulence factors can be considered to have a 'high virulence state'; an organism producing lower levels of virulence factors can be considered to have a 'low virulence state'. Virulence can be understood not only in terms of proximate causes— those specific traits of the pathogen that help make the host ill— but also by ultimate causes— the evolutionary pressures that lead to virulent traits occurring in a pathogen strain ("Virulence". Wikipedia: The Free
Encyclopedia. Wikimedia Foundation, Inc. 07 Mar 2015 Web. 27 May 2015
<http://en.wikipedia.org/wikiA^irulence>). In other words, the virulence state of, e.g., a bacterium, can be determined by a complex interplay between host and bacterial factors. In some contexts, virulence can be defined as an infection-induced loss of host fitness. The virulence of a microorganism may be assessed by any number of parameters, or combinations thereof, as known in the art. Non-limiting examples of parameters used to assess
microorganism virulence include adherence of the microorganism to cells or tissues of a subject (a host organism), colonization, invasion of cells or tissues, secretion of toxins resulting in death or damage of cells or tissues, formation of biofilms on cells or tissues, formation of biofilms on exogenous materials (i. e. , materials implanted temporarily or permanently in a subject), evasion or alteration of immune or other responses in the subject, resistance to antibiotics, scavenging of necessary nutrients or cofactors (e.g., iron), and/or motility. The virulence state of a microorganism such as P. aeruginosa is thus determined by a variety of factors, including genetic factors, induction of virulence factors and host responses, including specific and non-specific immune responses.
A plausible mechanism by which administration of an OprF antigen of the invention results in reduced mortality is the impact of the host immune response to the OprF antigen on the virulence state of the bacterium, possibly mediated in part by antibodies interfering with OprF function. More generally, factors affecting bacterial virulence can be broadly classified as virulence inhibitors. Example of a host-produced virulence inhibitor in the context of the current invention may include OprF-specific antibodies (see Fig. 1 for a schematic representation of potential effects of host immunization on P. aeruginosa virulence). In this regard, Wu et al. revealed one mechanism by which a prokaryotic organism can be directly signaled by immune activation in a eukaryotic host, by demonstrating that human interferon-γ binds to OprF protein in P. aeruginosa, resulting in the expression of a quorum-sensing dependent virulence determinant, the PA-I lectin (Wu et al, Recognition of Host Immune Activation by Pseudomonas aeruginosa, Science, 2005, 309:774-777). Additionally, Ding et
al. showed that host-produced IFN-γ increased mortality in a mouse model oiP. aeruginosa infection and further demonstrated that sera from OprF/I-immunized patients inhibited IFN- γ binding to OprF (Ding, B. et al, OprF/I vaccinated sera inhibit binding of human interferon-gamma to Pseudomonas aeruginosa, Vaccine, 2010, 28:4119-4122). These findings illustrate one mechanism by which OprF-specific immune responses may affect the virulence oiP. aeruginosa, and thus affect the outcome of an infection. Furthermore, P. aeruginosa infection exacerbates liver disease, in part by release of toxins such as pyocyanin, exotoxin A and endotoxin (LPS), leading to an increased risk of mortality in an already at- risk patient population (Chevaluppa, et al, International Journal of Infectious Diseases 14 (2010) e857-e867). Said hypothesis that the main use of the vaccination with an OprF antigen of the invention is the interference of the induced antibodies, i.e. blocking of the OprF function by antibodies induced by the OprF antigen vaccination. This hypothesis is novel and surprising but fits with the clinical observation as shown herein. The classical vaccine model of opsonophagocytosis (OPK) of the pathogen, here P. aeruginosa, does not explain the observed P. aeruginosa infections in the vaccinated and non-vaccinated groups (i.e. there was not difference observed). This interpretation allows new prevention and treatment strategies of which some are explored and defined herein. E.g. with this hypothesis it can be explained why the hepatobiliary disorder subjects are surprisingly benefiting from the vaccination as herein proposed.
Assessing effects of OprF antigen vaccination on the virulence of a P. aeruginosa can be carried out in vitro by, for example, quantifying the transcription of virulence factors or assessing other measures of virulence such as adherence or motility in the presence of host- produced factors. The host factors may be present, for example, in fluids of the immunized subject, such as, for example, blood, serum, sputum or urine. Assessing the interference of OprF function can be carried out in vitro by, for example, assessing the disruption of the OprF and OprF ligand (such as e.g. interferon gamma) interaction in an ELISA type assay.
Aspects of the invention provide methods for administering to a subject an OprF antigen. As used herein, an "antigen" refers to a molecule or a fragment or portion of a molecule that is immunogenic; in other words, stimulates an immune response; e.g., promotes the production of specific antibodies or proliferation and/or activation of effector cells in the subject. In some embodiments, the immune response is a humoral immune response; i.e., production of antibodies. In some embodiments, the immune response is a cell-mediated immune response. In some embodiments, the immune response is characterized by the
production and/or release of immune regulating molecules; for example, cytokines or chemokines. The presence of an immune response can be determined by any number of techniques of common usage in the art; for example assessment of the serum levels of antigen-specific antibodies as measured by an increase in Geometric Mean Titer (GMT) and compared with sera from placebo-treated subjects or pre-immune serum from the same patient to determine seroconversion in response to vaccination. Briefly, GMT can be assessed by an in vitro test which shows the levels of antigen specific antibodies in the sera of patients.
In some embodiments, the OprF antigen comprises or consists of a molecule that is able to stimulate the production of specific antibodies against OprF that are able to disrupt the binding of an OprF ligand to OprF (herein also generally referred to as "OprF antigen").
In further embodiments, the OprF antigen comprises or consists of the OprF polypeptide (SEQ ID NO: 1) or a fragment or a portion thereof. In some embodiments, the OprF antigen is a portion of a fusion protein that includes a portion of at least one additional protein. In some embodiments, the fusion protein is an OprF-OprI (OprF/I) fusion protein. OprF/I fusion proteins have been described, for example, in WO 2012/084272 and WO 2012/126879, herein incorporated by reference in their entirety. In some embodiments, the OprF/I fusion proteins comprise a portion of the P. aeruginosa outer membrane protein F (OprF, SEQ ID NO: 1) fused via the carboxy terminal end to a portion of the P. aeruginosa outer membrane protein I (Oprl, SEQ ID NO: 2). In some embodiments, the portion οΐΡ. aeruginosa OprF comprises amino acids 190-342 of P. aeruginosa OprF and amino acids 21- 83 οΐΡ. aeruginosa Oprl. In some embodiments, the OprF/I protein comprises an N-terminal tag for purification purposes, such as a His-tag, especially His6, preceded N-terminally by an alanine residue. In some embodiments, the N-terminal alanine residue is preceded N- terminally by a methionine residue. In a preferred embodiment, the N-terminal methionine residue is cleaved off during production of the OprF/I fusion protein. In some embodiments, the OprF/I fusion protein comprises or consists of the polypeptide provided by SEQ ID NO: 3.
In some embodiments, the OprF/I fusion protein is an immunogenic variant of the OprF/I fusion protein provided by SEQ ID NO: 3. An immunogenic variant may comprise one or more amino acid variations in the sequence of the OprF/I fusion protein, provided that the variant maintains at least the same or not less than 80%, more preferably 85%, even more preferably 90%, even more preferably 95%, most preferred 98% of the immunogenic activity of the original molecule. Immunogenic activity is preferably assessed by determination of the GMT, as described above, in the sera of immunized subjects compared with pre-immune sera
or sera from control (placebo) treated subjects. The amino acid variations may be mutations, such as, but not limited to, substitutions, insertions or deletions of residues, and may be made, for example, to enhance immunogenicity of the OprF antigen in the subject and/or the production of the OprF antigen (e.g. , by a recombinant cell). In some embodiments, the amino acid variations are conservative amino acid residue substitutions. As used herein, a "conservative amino acid substitution" is an amino acid substitution that does not alter the relative charge or size characteristics of the protein. Examples of conservative amino acid substitutions include the "conservatively modified variants" and "conservative sequence modifications" of polypeptides described in WO 2012/084272. In a preferred embodiment, the immunogenic variant retains the cysteine residues present at positions 18, 27, 33 and 47 of SEQ ID NO: 3.
In some embodiments, the OprF/I fusion protein comprises a variant polypeptide that is at least 80% (e.g., 85%, 90%, 95%, 96%, 97%, 98%, or 99%) identical to the OprF/I polypeptide provided by SEQ ID NO: 3, wherein the variant polypeptide maintains at least the same or not less than 90%, not less than 85%, or not less than 80% of the immunogenic activity of SEQ ID NO: 3, wherein the immunogenic activity is assessed as outlined above. The terms "identical" or "identity" in the context of two or more nucleic acid or polypeptide sequences, refer to two or more sequences or subsequences that are the same. Two sequences are "substantially identical" if two sequences have a specified percentage of amino acid residues or nucleotides that are the same (i. e., at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity) over a specified region or over the entire sequence, when compared and aligned for maximum correspondence over a comparison window, or designated region, as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Optionally, the identity exists over a region that is at least about 30 nucleotides (or 10 amino acids) in length, or more preferably over a region that is 60 to 150 or 600 or more nucleotides (or 20, 50, 200 or more amino acids) in length.
For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. Methods of alignment of sequences for comparison are well known in the art; for example, the local homology algorithm of Smith and Waterman (Comparison of biosequences (1981) Adv. Appl. Math. 2:482^189), the homology alignment algorithm of Needleman and Wunsch (A general method applicable to the search for similarities in the amino acid sequence of two proteins (1970) J. Mol. Biol. 48:443-53), the search for similarity method of Pearson and Lipman (Improved tools for biological sequence comparison (1988) Proc. Natl. Acad. Sci. USA 85 :2444-8), also computerized
implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the
Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr.,
Madison, WI) or manual alignment and visual inspection (see. e.g., Brent et al., Current Protocols in Molecular Biology, John Wiley & Sons, Inc. (Ringbou ed., 2003)). Two examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST 2.0 and BLAST algorithms, which are described in Altschul et al. (Gapped BLAST and PSI-BLAST: a new generation of protein database search programs (1997) Nuc. Acids Res. 25:3389-3402); and Altschul et al. (Basic local alignment search tool (1990) J. Mol. Biol. 215:403-410), respectively.
Any of the antigens described herein, such as the OprF/I fusion proteins, may be produced by any method known in the art. For example, the antigen may be recombinantly produced in a eukaryotic or prokaryotic cell, such as a tissue culture cell, a bacterial cell, or a fungal or yeast cell; or may be synthesized by conventional means. In a preferred
embodiment, the antigens are purified and processed as detailed in WO 2012/126879. Also within the scope of the invention are nucleic acids encoding OprF antigens of the invention. In some embodiments, the nucleic acids encode the OprF polypeptide (SEQ ID NO: 1) or an antigenic fragment thereof. In some embodiments, the nucleic acids encode the OprF/I fusion protein (SEQ ID NO: 3) or an antigenic fragment or immunogenic variant thereof. In some embodiments, the protein or polypeptide encoded by the nucleic acid is produced by cells of the subject (e.g., using transcriptional and/or translational machinery of the cells of the subject). In such embodiments, the nucleic acid may further encode additional features known in the art for expression of the encoded antigen.
The invention further encompasses OprF antigens as disclosed herein produced according to the following methods: recombinantly introducing an oligonucleotide encoding the OprF antigen into an expression vector, preferably pTRC99A, transforming a host cell, preferably E. coli, preferably E. coli strain XL1, and purifying and processing the resulting protein as detailed in WO 2012/126879, followed by storage at -80°C in PBS buffer. Briefly, the processing of the recombinantly -produced OprF/I protein comprises a reduction step with dithiothreitol (dtt) and a controlled re-oxidation step in the presence of L-cystine. The thus processed OprF antigen is further purified before storage and/or formulation.
An OprF antigen such as e.g. SEQ ID NO: 3 may be formulated e.g. as an injectable, such as for intramuscular or intravenous, preferably intramuscular administration, in a dose of 100 meg in a physiological salt solution (0.81% weight per volume) with or without aluminium hydroxide (400 meg).
Provided herein are methods for administering to a subject an OprF antigen. In some embodiments, the subject is a mammalian subject, such as a human, non-human primate, rodent, rabbit, sheep, dog, cat, horse, llama or cow. In some embodiments, the subject is a human subject, such as a patient.
In particular embodiments, the invention relates to the compounds, uses and methods wherein the subject to be treated has a hepatobiliary disorder, a medical history of a hepatobiliary disorder or an indication or indications associated with a hepatobiliary disorder and/or is, has been, or is at risk of being, an intensive care unit patient. In some embodiments, said subject is mechanically ventilated during at least part of a hospital or ICU stay. In other preferred embodiments, the subject to be treated is a burn patient.
In general, hepatobiliary disorders (liver diseases) include any disorder that disrupts the normal function of the liver, gall bladder, bile duct and/or bile of the subject. Examples of hepatobiliary disorders include, without limitation, viral hepatitis (A, B or C or other), non- viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, porphyria, Reye's syndrome, sarcoidosis, toxic hepatitis, type 1 glycogen storage disease, tyrosinemia, Wilson disease or a structural or other bile duct disorder. Whether a subject has a hepatobiliary disorder or an indication associated with a hepatobiliary disorder will be evident to one of skill in the art and may be determined by performing an assessment, such as any of the assessments described herein.
In some embodiments, the subject with a hepatobiliary disorder, medical history of a hepatobiliary disorder and/or an indication or indications associated with a hepatobiliary disorder and/or is, has been, or is at risk of being an intensive care unit patient and/or a burn patient additionally has no previous history of nervous system disorders.
In some embodiments, an indication associated with a hepatobiliary disorder may refer to the result of a clinical assessment, such as the level of a protein found in the serum, that is outside (above or below) the range of a result considered to be normal. As used herein, "clinically relevant deviation" or "clinically relevant levels" or "clinically relevant elevation" refers to a result of an assessment that is outside the range of a result considered to be normal and may indicate the subject has or is at risk of having a hepatobiliary disorder.
In some embodiments, the methods include testing a subject for a hepatobiliary disorder or indication associated with a hepatobiliary disorder. In some embodiments, the subject may be tested for a hepatobiliary disorder or one or more indications associated with a hepatobiliary disorder prior to administration of the OprF antigen. Any assessment associated with or indicative of a hepatobiliary disorder or the dysfunction of the liver, gallbladder and/or bile duct may be compatible with the methods described herein. Examples of assessments include, without limitation, examining the medical history of the subject, performing a physical examination of the subject, measuring the level of one or more liver proteins in a serum sample from the subject, measuring hepatic excretory function, performing ultrasonography and/or computed tomography scanning of the liver, performing angiography and performing a liver biopsy. In some embodiments, the presence of liver proteins in a serum sample indicates the subject has a hepatobiliary disorder. In some embodiments, the presence of one or more liver proteins at a particular concentration (e.g., a concentration beyond the range considered normal) indicates that the subject has a hepatobiliary disorder. In some embodiments, the liver protein is serum glutamic pyruvic transaminase (SGPT; also referred to as alanine aminotransferase), serum glutamic oxaloacetic transaminase (SGOT; also referred to as aspartate aminotransferase), bilirubin, γ- glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase and/or albumin.
In general, a Respiratory, Thoracic or Mediastinal (RTM) disorder is any disorder that affects the upper or lower respiratory tract and associated structures or any organs, systems, tissues or cells in the anatomical region of the thorax or mediastinum. RTM disorders may include, for example, any of the following diseases, symptoms or syndromes: acquired diaphragmatic eventration, pleural disorder, acute pulmonary edema, pleural effusion, acute respiratory distress syndrome, pleural fibrosis, acute respiratory failure, pleurisy, alveolitis allergic, pleuritic pain, apnea, pneumomediastinum, asphyxia, pneumonia aspiration, aspiration, pneumonitis, asthma, pneumothorax, asthma late onset, productive cough, pleural effusion, atelectasis, prolonged expiration, bronchial disorder, pulmonary alveolar hemorrhage, bronchial fistula, pulmonary arterial hypertension, bronchial hyperreactivity, pulmonary cavitation, bronchial obstruction, pulmonary congestion, bronchiectasis, pulmonary embolism, bronchitis chronic, pulmonary fibrosis, bronchopleural fistula, pulmonary hemorrhage, bronchospasm, pulmonary hypertension, Cheyne-Stokes respiration, pulmonary mass, chronic obstructive pulmonary disease, pulmonary necrosis, chronic respiratory disease, pulmonary edema, chronic respiratory failure, rales, cough, respiratory acidosis, dysphonia, respiratory alkalosis, dyspnea, respiratory arrest, dyspnea exertional,
respiratory disorder, dyspnea paroxysmal nocturnal, respiratory distress, emphysema, respiratory failure, epiglottic edema, respiratory fatigue, epistaxis, respiratory gas exchange disorder, hemoptysis, respiratory tract inflammation, haemothorax, respiratory tract edema, hiccups, restrictive pulmonary disease, hydrothorax, rhinitis allergic, hypercapnia, rhonchi, hyperventilation, sleep apnea syndrome, hypoventilation, sneezing, hypoxia, sputum discolored, idiopathic pulmonary fibrosis, sputum increased, increased viscosity of bronchial secretion, status asthmaticus, interstitial lung disease, stridor, laryngeal oedema, tachypnea, lower respiratory tract inflammation, tonsillar hemorrhage, lung consolidation,
tracheomalacia, lung disorder, upper airway resistance syndrome, lung infiltration, wheezing, lupus pneumonitis, mediastinal shift, obstructive airways disorder, oropharyngeal pain, oropharyngeal spasm, orthopnea, pharyngeal hemorrhage, pharyngeal edema, pickwickian syndrome, pleural calcification, laryngospasm, pulmonary microemboli, neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and organizing pneumonia.
In some embodiments, the methods include testing a subject for RTM disorders. In some embodiments, RTM disorders can be assessed as deemed appropriate by the attending medical professional. In some embodiments, the subject may be tested for an RTM disorder prior to administration of the OprF antigen. Any assessment associated with or indicative of an RTM may be compatible with the methods described herein. Examples of assessments include, without limitation, examining the medical history of the subject, performing a physical examination of the subject, X-ray imaging, chest auscultation, palpation, magnetic resonance imaging (MRI), electrocardiogram (ECG), echocardiogram, pulse oxiometry, computerized axial tomography (CAT) scan, lung function tests, e.g., spirometry, and blood testing. The blood tests can assess, for example, arterial blood gas concentration, levels of indicator hormones such as atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP) or may also include, e.g. for the purposes of diagnostic elimination, other
complementary blood test assessments for kidney or thyroid function and total blood count.
In some embodiments, the subject is an ICU patient and/or a burn patient, has been an ICU patient and/or a burn patient, or is at risk of being an ICU patient and/or burn patient. In general, an ICU patient means that the subject has a medical condition requiring the support of an intensive care unit of a medical facility. In some embodiments, the subject may have a severe and/or life-threatening medical condition and/or injuries. Whether a subject is an ICU patient and/or a burn patient will be evident to one of skill in the art. In some embodiments, the subject has been an ICU patient, for example the subject was an ICU patient within the
previous 1 day to 6 months prior to administration of any of the OprF antigens described herein. In some embodiments, the subject was an ICU patient within at least the previous 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 25, 26, 27, 28, 29, 30 days, or at least 1, 2, 3, 4, 5, 6 months prior to administration of any of the OprF antigens described herein.
In some embodiments, the subject is or has been mechanically ventilated, for example using a positive pressure ventilator or a negative pressure ventilator.
In some embodiments, the subject is at risk of being an ICU patient. In some embodiments, the subject has a disease, disorder, or injury that has the risk of progressing to a stage that requires the support of an intensive care unit of a medical facility. In some embodiments, the subject may participate in activities that put the subject at risk of severe bodily injury that may require the support of an intensive care unit of a medical facility. Examples of activities that may put the subject at risk of severe injury include, without limitation, extreme sports, such as base jumping, bungee jumping, gliding, hang gliding, high wire, ski jumping, sky diving, sky surfing, sky flying, indoor climbing, adventure racing, aggressive inline skating, BMX, caving, extreme motocross, extreme skiing, freestyle skiing, land and ice yachting, mountain biking, mountain boarding, outdoor climbing, sandboarding, skateboarding, snowboarding, snowmobiling, speed biking, speed skiing, scootering, barefoot waterskiing, cliff diving, free-diving, jet skiing, open water swimming, powerboat racing, round the world yacht racing, scuba diving, snorkeling, speedsailing, surfing, wakeboarding, Whitewater kayaking, windsurfing. In some embodiments, any of the OprF antigens described herein may be administered to the subject prior to participation in an activity that may put the subject at risk of being an intensive care unit patient.
Alternatively or in addition to the subject being, having been, or at risk of being an intensive care unit patient, the subject may be, have been, or be at risk of being a burn patient. In some embodiments, the subject has or is at risk of sustaining a burn that requires support from a specialized burn facility. The severity of burns is generally classified based on the depth of skin and tissues affected by the burn. The burn or burns may be first-degree, second- degree, third-degree, fourth-degree, or a combination thereof. Whether a subject is a burn patient will be evident to one of skill in the art. In some embodiments, the subject has been a burn patient, for example the subject was a burn patient within the previous 1 day to 6 months prior to administration of any of the OprF antigens described herein. In some embodiments, the subject was a burn patient within at least the previous 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12,
13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 25, 26, 27, 28, 29, 30 days, or at least 1, 2, 3, 4, 5, 6 months prior to administration of any of the OprF antigens described herein.
In some embodiments, the subject is at risk of being a burn patient. In some embodiments, the subject may participate in activities that put the subject at risk of sustaining a burn, for example a burn that may require the support of a specialized burn facility. In some embodiments, the subject has an occupation that may put the subject at risk of sustaining a burn, for example, the subject is a firefighter, works in food preparation, or works with chemicals. In some embodiments, any of the OprF antigens described herein may be administered to the subject prior to participation in an activity or occupation that may put the subject at risk of being a burn patient.
In some embodiments, the subject at risk of becoming an ICU patient or a burn patient is elderly, immunocompromised, catheterized, undergoing surgery, a cancer patient, an HIV- infected patient or a cystic fibrosis patient and, as such, particularly at high risk for P.
aeruginosa infection. In some embodiments, these subjects may be administered any of the OprF antigens described herein at any time before admission or imminent admission into the ICU.
In some embodiments, the OprF antigen is administered to the subject once. In some embodiments, the OprF antigen is administered to the subject more than once. In some embodiments the OprF antigen is administered to the subject on at least 2, 3, 4, 5, 6, 7, 8, 9, 10 or more occasions. Any administration of the OprF antigen that is administered after the first administration may be referred to as a "booster".
In some embodiments, a subsequent second administration of the OprF antigen is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24,2 5, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or at least 60 days after the first administration of the OprF antigen (preferred for the second administration is 7 days after the first administration). In some embodiments, a subsequent (subsequent to the second and any further administrations) administration of the OprF antigen is administered at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24,2 5, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, or at least 60 days after the prior administration of the OprF antigen.
Determining whether a subject is in need of one or more additional administrations of the OprF antigen will be evaluated according to one of ordinary skill in the art.
The compounds, uses and methods described herein may result in a reduction of the mortality in a subject being administered the OprF antigen. As used herein, "mortality" means death of a subject. "Survival" means non-death of a subject. The "overall survival" ("OS") of the subjects in a given group or "survival" can also be expressed as a percentage of survivors in the group at a given timepoint; for example, day 28 or day 90 survival. The survival is the inverse of the mortality. Any expression of mortality can also be expressed inversely as survival.
The term "increased survival" or "increased overall survival" refers to a comparison of the survival of two different patient groups, especially to the difference in survival between treated and placebo patient groups, particularly at a given timepoint. An example of increased survival in the current invention is the survival in subjects who are administered an OprF antigen (e.g., a treated subject group) minus the survival in subjects who do not receive the treatment (i.e., a placebo control group) and in case where the survival in the treated group is higher than in the placebo group the difference is > 0% and thus the overall survival or survival is increased. In particular, e.g.:
Increased survival at day 28 (d28) if the survival of treated group at d28 minus survival of placebo group at d28 > 0%;
or,
Increased survival of the subject (at e.g. d28) if the ratio of the mortality of treated subjects (at e.g. d28) versus the mortality of placebo controlled subjects (at e.g. d28) times one hundred (xlOO) > 100; i.e.,
(mortality of treated subjects [at e.g. d28]/mortality of placebo controlled subjects [at e.g. d28]) x 100 >100
As used herein, the term "mortality" refers to a measure of the number of deaths in a target population under certain conditions (e.g., a subject with a past or ongoing hepatobiliary disorder that is an intensive care unit patient and that is administered the OprF/I fusion protein set forth as SEQ ID NO: 3) per unit time (e.g., 28 days from first administration of the OprF/I fusion protein set forth as SEQ ID NO: 3), expressed as a percentage. Aspects of the invention describe e.g., the mortality; i.e., the number of deaths in the population of subjects within the period of the first treatment administration with e.g., the OprF/I fusion protein (day 0) until 28 days later (day 28; referred to herein as day 28 mortality) or until 90 days later (day 90; referred to herein as day 90 mortality) expressed as a percentage.
As used herein, the term "reduced mortality" is defined as the difference in the mortality of a treated group in comparison with the mortality in a non-treated group (e.g., a placebo group), particularly at a given timepoint. More specifically, reduced mortality is
when the difference in the mortality of placebo controlled patients compared with the mortality of drug treated patients (e.g., in the OprF/I treated patient group) is greater than 0%. For example: Mortality at day 28 (d28) is reduced if the mortality of placebo group at d28 minus mortality of treated group at d28 > 0%;
or,
Reduced mortality of the subject (at e.g. d28) is true if the ratio of the mortality of treated subjects (at e.g. d28) versus the mortality of placebo controlled subjects (at e.g. d28) times one hundred (xlOO) < 100; i.e., (mortality of treated subjects [at e.g. d28]/mortality of placebo controlled subjects [at e.g. d28]) x 100 < 100
In some embodiments, the mortality of the subject being administered the OprF antigen is reduced relative to the mortality of a subject who is not administered the OprF antigen; i.e., the placebo control. In some embodiments, the mortality of the subject being administered the OprF antigen is reduced compared to a subject that did not receive treatment (i.e., received a placebo control) at day 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, or at least 90 days after administration. In some embodiments, the mortality of the subject being administered the OprF antigen is reduced compared to a subject that did not receive treatment at day 28 after administration. In some embodiments, the mortality of the subject being administered the OprF antigen is reduced compared to a subject that did not receive treatment at day 90 after administration. In some embodiments, the mortality of the subject being administered the OprF antigen is reduced by at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 1 1%, 12%, 13%, 14%, 15%, 16%, 17%, 18%, 19%, 20%, 21%, 22%, 23%, 24%, 25%, 26%, 27%, 28%, 29%, 30%, 31%, 32%, 33%, 34%, 35%, 36%, 37%, 38%, 39%, 40%, 41%, 42%, 43%, 44%, 45%, 46%, 47%, 48%, 49%, 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59% or at least 60% compared to a subject that did not receive treatment. In a preferred embodiment, the reduced mortality of the subject being administered the OprF antigen is at least 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94 or at least 95 compared to a subject that did not receive treatment.
As used herein, the term "probability of dying" is defined as the probability of dying in a particular situation for a subject. E.g. the particular situation is a hepatobiliary subject being in, or shortly before being admitted to an ICU, benefiting from the treatment and preventative strategies disclosed and claimed herein and has a reduced probability of dying as shown herein. E.g. the difference in the mortality of a treated group in comparison with the mortality of a non-treated group (e.g., a placebo group), particularly at a given timepoint after vaccination, e.g. 21 days after the second vaccination, is reduced and thus for the individual subject the probability of dying has been reduced, i.e. the outlook of survival for the individual subject has improved. The reduced probability of dying of the subject correlates with the degree of reduction of observed mortality, i.e. the higher the reduced mortality, the lower the probability of dying of the subject. This observation is particularly surprising as mortality is reduced, but the the overall Pseudomonas aeruginosa infection rate has not been reduced despite the vaccination against Pseudomonas aeruginosa (see also supra for a possible explanation of the mechanism of action).
Any of the methods or uses described herein may be for the treatment or prevention of a P. aeruginosa infection and/or a symptom thereof in a subject. As used herein, the terms "treat" and "treating" shall mean the administration of an OprF antigen to a subject to cure, ameliorate, prevent, reduce, or delay the onset of the symptoms, complications, pathologies, sequelae or biochemical indicia of a disease or infection (such as e.g., prevent or delay infections in subjects), alleviating the symptoms or arresting or inhibiting further
development of the disease, infection, condition, or disorder. Treatment may be prophylactic, i.e. prevention or delay of onset of the disease, or prevention or reduction of manifestations of clinical or subclinical symptoms thereof or therapeutic, i.e. suppression or alleviation of symptoms after the manifestation of the disease. The term "prevention of as used herein shall mean "protection from" in the sense that the severity of disease, in the case of infection, is reduced. An OprF antigen that is administered to a subject prophylactically (e.g., including prior to infection but generally also including any administration where the infection is not severely affecting the health of the subject) may be referred to as a vaccine. In preferred embodiments, the subject has or is at risk of having hepatobiliary disorders, for example having one or more indicia of hepatobiliary disorders, as otherwise herein described. In a preferred embodiment, the subject has no history of or existing RTM disorders.
In some embodiments, the subject has a P. aeruginosa infection. P. aeruginosa may cause any of a variety of types of infections in a subject, for example, the infection may be of the respiratory tract (e.g., pneumonia), bloodstream (bacteremia), heart (endocarditis), central
nervous system (e.g., meningitis, brain abscess), ear (e.g., otitis externa, otitis media), eye (e.g., bacterial keratitis, endophthalmitis), bones and joints (e.g., osteomyelitis),
gastrointestinal tract (e.g., diarrhea, enteritis, entercolitis), urinary tract (e.g., cystitis, pyelonephritis) or skin (e.g., ecthyma gangrenosum). In general, P. aeruginosa infections may be classified as acute or chronic infections. Acute P. aeruginosa infections are characterized as rapidly spreading, causing tissue damage and/or sepsis, and may be associated with a high mortality rate. Chronic P. aeruginosa infections are characterized as persisting for weeks, months or years. In some embodiments, the subject has an acute P. aeruginosa infection. In some embodiments, the subject has fulminant P. aeruginosa infection, such as, for example, bacterial keratitis. "Fulminant" as applied to an infection and as used herein shall mean an aggressive acute disease condition; i.e., one which is characterized by rapid onset of a new infection or sudden and rapid worsening of an existing infection and which is especially severe and/or life-threatening. A fulminant infection can furthermore arise from chronic infections. In some embodiments, the subject has a chronic P. aeruginosa infection. In some embodiments, the P. aeruginosa infection may be the result of or caused by the subject having been mechanically ventilated.
Any of the OprF antigens described herein may be administered to a subject with, prior to, or after administration of an adjuvant. An adjuvant is an agent that enhances a response in a subject, such as an immune response, to an antigen or other molecule. In some embodiments, an adjuvant may stabilize an antigen or other molecule. Determining whether an OprF antigen is administered with an adjuvant depends on various factors (e.g., type and extent of response desired) and will be evident to one of skill in the art. Examples of adjuvants may include, without limitation, alum, IC31 ®, aluminum hydroxide, aluminum phosphate, calcium phosphate hydroxide, paraffin oil, killed bacteria, bacterial toxins, toxoids, subunits of bacteria, squalene, detergents, IL-1, IL-2, IL-12, TLR9 agonists and combinations such as Freund's complete adjuvant and Freund's incomplete adjuvant. In some embodiments, the OprF antigens are administered with aluminum hydroxide. In some embodiments, the OprF antigens are administered without adjuvant.
Any of the OprF antigens described herein may be administered to a subject in an effective amount. As used herein, an "effective amount" of an OprF antigen is any amount that results in a desired response or outcome in a subject, such as those described herein, including but not limited to reduction of mortality or risk of mortality. In some embodiments, the OprF antigen may be formulated for administration in a pharmaceutical composition. The term "pharmaceutical composition" as used herein means a product that results from the
mixing or combining of one or more active ingredients and includes both fixed and non- fixed combinations of the one or more active ingredient(s). The term "fixed combination" means that the active ingredients, e.g., the OprF antigen and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g., the antigen and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides
therapeutically effective levels of the two or more compounds in the body of the patient. A pharmaceutical composition as used herein can also mean a vaccine or vaccine composition.
Pharmaceutical compositions of the invention, including vaccines, can be prepared in accordance with methods well-known and routinely practiced in the art (see e.g., Remington: The Science and Practice of Pharmacy, Mack Publishing Co. 20th ed. 2000; and Ingredients of Vaccines - Fact Sheet from the Centers for Disease Control and Prevention).
Pharmaceutical compositions are preferably manufactured under GMP conditions. Typically, a pharmaceutically effective dose of the OprF antigen is employed in the pharmaceutical composition of the invention. The OprF antigen is formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Dosage regimens are adjusted to provide the optimal desired response (e.g., the therapeutic or prophylactic response).
Dosage levels of the active ingredients in the pharmaceutical compositions of the present invention can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired pharmaceutical response for a particular subject, composition, and mode of administration, without being toxic to the subject. The selected dosage level depends upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the subject being treated and like factors.
A physician, veterinarian or other trained practitioner can administer doses of the
OprF antigen of the invention employed in the pharmaceutical composition at levels lower than that required to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, effective doses of the compositions of the present invention for the prophylactic and therapeutic treatment of groups of people as
described herein vary depending upon many different factors, including means of administration, target site, physiological state of the patient, whether the patient is a human or an animal, other medications administered and whether treatment is prophylactic or therapeutic. Treatment dosages need to be titrated to optimize safety and efficacy.
For systemic administration of an OprF/I agent such as a vaccine comprising an
OprF/I fusion protein, the doses range between 10 and 1000 meg, 50 to 500 meg, 100 to 200 meg, especially 100 meg. An exemplary treatment regime entails systemic administration; e.g., twice or once for a vaccine. In a preferred embodiment, the vaccine is administered twice. As used herein, "systemic administration" includes such routes of delivery as subcutaneous, intramuscular, intradermal, intranasal, intravaginal or intrarectal, preferably intramuscular. An exemplary treatment regime entails systemic administration at day 0 and day 7 for the vaccine consisting of 100-200 meg SEQ ID NO: 3 and approximately 0.9% weight per volume NaCl in water or PBS, and optionally containing alum, preferably without alum. Herein, the OprF/I fusion protein according to SEQ ID NO:3, as well as adjuvanted or non-adjuvanted formulations comprising the OprF/I fusion protein according to SEQ ID
NO:3, are also referred to as "IC43". Day 0 shall be defined as the day on which the subject receives the first vaccination which shall preferably be the day of admittance to the hospital or ICU or, alternatively, the day on which the condition of the patient necessitates mechanical ventilation.
The invention is not limited in its application to the details of construction and the arrangement of components set forth in the following description or illustrated in the drawings. The invention is capable of other embodiments and of being practiced or of being carried out in various ways. Also, the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of "including", "comprising", or "having", "containing", "involving" and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.
Unless otherwise defined herein, scientific and technical terms used in connection with the present disclosure shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. The methods and techniques of the present disclosure are generally performed according to conventional methods well- known in the art. Generally, nomenclature used in connection with, and techniques of biochemistry, enzymology, molecular and cellular biology, microbiology, virology, cell or tissue culture, genetics and protein and nucleic acid chemistry described herein are those
well-known and commonly used in the art. The methods and techniques of the present disclosure are generally performed according to conventional methods well-known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification unless otherwise indicated.
The present invention is further illustrated by the following Examples, which in no way should be construed as further limiting. The entire contents of all of the references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated by reference, in particular for the teaching that is referenced hereinabove. However, the citation of any reference is not intended to be an admission that the reference is prior art.
BRIEF DESCRIPTION OF THE DRAWINGS
The accompanying drawings are not intended to be drawn to scale. The Figures are illustrative only and are not required for enablement of the disclosure. For purposes of clarity, not every component may be labeled in every drawing. In the drawings:
Figure 1 shows a schematic representation of potential effects of virulence inhibitors, which may include antibodies produced in response to IC43 as shown, on the virulence of Pseudomonas and potential outcomes of these effects during an infection. The image represents an OprF protein assembled in the outer membrane ΐΡ. aeruginosa.
Figure 2 shows the percent survival over time of all subjects in clinical trial IC43-201 following the administration of three different formulations of IC43 or placebo. The treatments included the following: 100 micrograms of the OprF/I fusion protein without adjuvant (IC43 100 meg w/o, 97 subjects), 100 micrograms of the OprF/I fusion protein with alum (IC43 100 meg, 107 subjects), 200 micrograms of the OprF/I fusion protein with alum (IC43 200 meg, 100 subjects) and alum only (Placebo, 98 subjects). Subjects were vaccinated on days 0 and 7 and survival was monitored. Censoring is indicated by the presence of crosses.
Figure 3 shows the percent survival over time of the subset of subjects in the IC43- 201 trial who presented at the start of the trial with a previous medical history of
hepatobiliary disorders compared with those subjects with no such history. The data from all three IC43 treatment groups (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) were combined and the overall survival of the two populations is shown (IC43 MedHist
Hepatobiliary disorders YES and IC43 MedHist Hepatobiliary disorders NO) and compared
with the placebo-treated groups (Placebo MedHist Hepatobiliary disorders YES and Placebo MedHist Hepatobiliary disorders NO). (The p-value is with respect to the IC43-treated versus placebo groups of subjects with a medical history of hepatobiliary disorders and was calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
Figure 4 shows the survival over time of the subset of patients in the IC43-201 trial with a previous medical history of hepatobiliary disease as in Figure 3, with the results of each of the IC43 formulations shown separately. (The p-values are with respect to the IC43- treated versus placebo groups of subjects with a medical history of hepatobiliary disorders and were calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
Figure 4A shows the survival plot of subjects in the IC43-201 trial following day 0 and day 7 administration of 100 micrograms of non-adjuvanted OprF/I fusion protein or placebo.
Figure 4B shows the survival plot of subjects in the IC43-201 trial following administration of 100 micrograms of OprF/I fusion protein with adjuvant or placebo.
Figure 4C shows the survival plot of subjects in the IC43-201 trial following administration of 200 micrograms of OprF/I fusion protein with adjuvant or placebo.
Figure 5 shows the survival over time of the subset of patients in the IC43-201 trial with clinically-relevant elevations of serum levels of two liver enzymes: serum glutamic oxaloacetic transaminase (SGOT) and serum glutamic pyruvic transaminase (SGPT) following IC43 or placebo administration. (P-values are with respect to the IC43-treated versus placebo groups in subjects with elevated SGOT or SGPT and were calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
Figure 5A shows the survival over time of the subset of subjects in the IC43-201 trial with clinically relevant (CR) SGOT levels at the start of the trial compared with those subjects with non-clinically relevant (NCR) SGOT levels following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo. The data from all three IC43 treatments (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) are combined and the overall survival of the two populations is shown (IC43 SGOT.V0 CR and IC43 SGOT.V0 NCR) and compared with the placebo-treated groups (Placebo SGOT.V0 CR and Placebo SGOT.V0 NCR). Subjects with a clinically relevant deviation of SGOT at base-line in the IC43 and placebo groups are shown (lighter solid lines and dashed lines, respectively) and patients without clinically relevant deviation of SGOT at base-line in the IC43 -treated and
placebo cohorts are shown (darker solid lines and dashed lines, respectively). The number of patients in each group is indicated in the legend.
Figure 5B shows the survival over time of the subset of subjects in the IC43-201 trial having CR SGPT levels at the start of the trial compared with those subjects with NCR SGPT levels following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo. The data from all three IC43 treatments (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) are combined and the survival over time of the two populations is shown (IC43 SGPT.VO CR and IC43 SGPT.VO NCR) and compared with the placebo-treated groups (Placebo SGPT.VO CR and Placebo SGPT.VO NCR). Subjects with clinically relevant deviation of SGPT at BL in the IC43 and placebo cohorts are shown (lighter solid lines and dashed lines, respectively) and patients without clinically relevant deviation of SGPT at BL in the IC43-treated and placebo cohorts are shown (darker solid lines and dashed lines, respectively). The number of patients in each group is indicated in the legend.
Figure 5C shows the overlap between three patient subsets selected from all IC43- and placebo-treated patients in the IC43-201 trial: those with a previous medical history of hepatobiliary disorders (77), those with clinically-relevant SGOT serum levels (69) and those with clinically relevant SGPT serum levels (48).
Figure 6A shows the survival over time of the subset of subjects in the IC43-201 trial with an abnormal hepatic physical examination as measured at baseline compared to subjects with a normal hepatic physical examination following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo. Data from all three IC43 -treated groups (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) are combined and the survival over time of the two populations is shown (IC43 PhyExam Hepatic ABNORMAL and IC43 PhyExam Hepatic NORMAL) and compared with the placebo-treated groups (Placebo PhyExam Hepatic ABNORMAL and Placebo PhyExam Hepatic NORMAL). (The p-value is with respect to the IC43 -treated and placebo groups with an abnormal hepatic examination and was calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
Figure 6B shows the overlap of two patient subsets from the IC43-201 trial: those with an abnormal hepatic physical examination (88 in total) and those with a previous medical history of hepatobiliary disorders (77 in total).
Figure 7 lists the types of hepatobiliary disorders identified in patients taking part in the IC43-201 clinical trial and the numbers of patients with the respective disorder. (NEC = not elsewhere classified.)
Figure 8A shows the survival over time of the subset of subjects in the IC43-201 trial with no medical history of respiratory, thoracic or mediastinal disorders at baseline compared to subjects with a medical history of respiratory, thoracic or mediastinal disorders following day 0 and day 7 administration of the OprF/I fusion protein (IC43) or placebo. Data from all three IC43 -treated groups (IC43 100 meg w/o, IC43 100 meg and IC43 200 meg) are combined and the survival over time of the two populations is shown (IC43 w/o RTM and IC43 with RTM) and compared with the placebo-treated groups (Placebo w/o RTM and Placebo with RTM). (The p-value is with respect to the IC43 -treated and placebo groups with no medical history of respiratory, thoracic or mediastinal disorders and was calculated using the Log-Rank test.) Censoring is indicated by the presence of crosses.
Figure 8B shows the overlap of two patient subsets in the IC43-201 trial: those with no medical history of respiratory, thoracic or mediastinal disorders (153 in total) and those with a previous medical history of hepatobiliary disorders (77 in total).
SEQUENCES
The amino acid sequence of OprF is provided by SEQ ID NO: 1 :
1 MKLKNTLGW I GS LVAASAM NAFAQGQNSV E I EAFGKRYF TDSVRNMKNA DLYGGS I GYF
61 LTDDVELALS YGEYHDVRGT YETGNKKVHG NLTS LDAI YH FGTPGVGLRP YVSAGLAHQN
12 1 I TNINSDSQG RQQMTMANI G AGLKYYFTEN FFAKAS LDGQ YGLEKRDNGH QGEWMAGLGV
18 1 GFNFGGSKAA PAPEPVADVC SD SDNDGVCD NVDKCPDTPA NVTVDANGCP AVAEWRVQL
24 1 DVKFDFDKSK VKENSYAD I K NLADFMKQYP ST STTVEGHT D SVGTDAYNQ KLSERRANAV
30 1 RDVLVNEYGV EGGRVNAVGY GE SRPVADNA TAEGRAINRR VEAEVEAEAK
The amino acid sequence of Oprl is provided by SEQ ID NO: 2:
1 MNNVLKFSAL ALAAVLATGC S SHSKETEAR LTATEDAAAR AQARADEAYR KADEALGAAQ 61 KAQQTADEAN ERALRMLEKA SRK
The amino acid sequence of the OprF/I fusion protein (IC43) is provided by SEQ ID NO: 3
1 AHHHHHHAPA PEPVADVC SD SDNDGVCDNV DKCPDTPANV TVDANGCPAV AEVVRVQLDV 61 KFDFDKSKVK ENSYAD I KNL ADFMKQYP S T STTVEGHTDS VGTDAYNQKL SERRANAVRD 12 1 VLVNEYGVEG GRVNAVGYGE SRPVADNATA EGRAINRRVE S SHSKETEAR LTATEDAAAR 18 1 AQARADEAYR KADEALGAAQ KAQQTADEAN ERALRMLEKA SRK
EXAMPLES
The following Examples summarize a subset of available data compiled from the results of two clinical trials, namely IC43-201 and IC43-202, wherein patients were vaccinated with IC43 or placebo. The differences between the inclusion and exclusion criteria and the primary and secondary endpoints of the two trials are shown in Tables Al and A2, respectively.
Table Al. Patient Inclusion and Exclusion criteria in clinical trials IC43-201 and IC43-202.
Clinical trial IC43-201 Clinical trial IC43-202
Patient Inclusion Criteria Patient Inclusion Criteria
Male or female patients admitted to an ICU with need Male or female patients admitted to an ICU with need for for mechanical ventilation for at least 48 hours aged 18 mechanical ventilation for at least 48 hours aged 18 to 80 to 80 years at Visit 0 years at Visit 0
Patients who, as determined by the investigator, have a Patients who, as determined by the investigator, have a high probability of survival for at least 48 hours high probability of survival for at least 48 hours.
In females, either childbearing potential terminated by Either no childbearing potential (terminated by surgery or surgery or 1 year post-menopausal, or a negative serum 1 year post-menopausal) or negative pregnancy test and pregnancy test and the willingness of practicing a the willingness of practicing a reliable method of reliable methods of contraception contraception.
Written informed consent according to the national Written informed consent (e.g., by the patient or his/her regulations (e.g. by the patient or legal representative) legally authorized representative) or waiver according to the national regulations
Patient Exclusion Criteria Patient Exclusion Criteria
Known use of any other investigational or non- Known use of any other investigational or non-registered registered drug (except the study vaccine) within 30 drug (except the study vaccine) within 30 days prior to days prior to IC43 vaccination at Visit 0 IC43 vaccination at Visit 0 or during the study
Pregnancy (positive pregnancy test at study entry), Pregnancy (positive pregnancy test at Visit 0 or Visit 1), lactation lactation
Persons who have been committed involuntarily to an Persons who have been committed involuntarily to an institution, e.g. mental health facility or prison, will not institution, e.g. mental health facility or prison participate in the study SOFA < 4 at Visit 0
Patients receiving immunosuppressive therapy after Patients < 6 months post organ transplantation transplantation whose immunosuppressive therapy has
not been stable within the last two months (either dose Severe thrombocytopenia (<50,000/μ1) or other escalation or change in medication) coagulopathy which in the opinion of the investigator makes the patient unsuitable for intramuscular injection
Known family history of congenital or hereditary
immunodeficiency Expected plasmapheresis or immunoadsorption during study period (at Visit 0)
Infection with HIV (a negative test result within 30
days before screening is acceptable Readmission to ICU during the current total hospital stay on Day 0
Severe thrombocytopenia or other coagulopathy which
in the opinion of the investigator makes the patient Patients admitted to ICU within 2 days after surgery unsuitable for intramuscular injection Patients admitted to ICU due to trauma
Any condition which in the opinion of the investigator Elective surgery until Day 28 after first vaccination makes the patient unsuitable for inclusion
Table A2. Primary and Secondary endpoints in clinical trials IC43-201 and IC43-202.
Clinical trial IC43-201 Clinical trial IC43-202
Primary endpoint Primary endpoint
To investigate the immunogenicity of IC43 in ■ Day 28 all cause mortality in patients receiving IC43 or placebo mechanically ventilated ICU patients at day 14
after first vaccination
Secondary endpoints Secondary endpoints
To investigate the immunogenicity of IC43 in Efficacy endpoints:
mechanically ventilated ICU patients at day 7 and
Day 14, 56 and 90 all cause mortality in patients receiving IC43 in biweekly intervals after day 14 until ICU
or placebo
discharge and day 90
Day 28, 56 and 90 all cause mortality in patients surviving Day
To investigate the safety and tolerability of IC43
14 and receiving IC43 or placebo
during a period of up to 90 days after the first
vaccination Day 14, 28, 56 and 90 all cause mortality in patients surviving
Day 3 and receiving IC43 or placebo
To estimate the rate of invasive Pseudomonas
aeruginosa (P. aeruginosa) infection in (ICU) Overall survival in all patients and in patients surviving Day 14 patients receiving IC43 vaccination versus Day 28 all cause mortality in patients with documented medical placebo during ICU stay history of a hepatobiliary disorder receiving IC43 or placebo
To analyze and compare the incidence rates of Overall survival in patients with documented medical history of colonization by P. aeruginosa in ICU patients a hepatobiliary disorder
receiving IC43 vaccination versus placebo during
ICU stay Sepsis related-mortality at Day 14, 28, 56 and 90 in patients receiving IC43 or placebo
To analyze the impact of IC43 vaccination on
organ function Sepsis related survival in patients receiving IC43 or placebo
To determine supportive immunological In-ICU and in-hospital mortality in patients receiving IC43 or endpoints (OP , OPA) placebo until day 14, 28, 56, 90, 180
Percentage of patients with invasive infection with P.
aeruginosa, such as bacteremia (determined as positive blood culture) or P. aeruginosa urinary tract infection in patients receiving IC43 or placebo within 28 and 56, 90 and 180 days after first vaccination
Percentage of patients with P. aeruginosa respiratory tract infection (pneumonia or tracheobronchitis) or P. aeruginosa respiratory tract colonization in patients receiving IC43 or placebo within 28 and 56, 90 and 180 days after first vaccination
Organ function (Sequential Organ Failure Assessment [SOFA] scores) in patients receiving IC43 or placebo during ICU stay
Length of ICU stay in patients receiving IC43 or placebo
Immunogenicity endpoints:
■ Immunogenicity at Day 7, 14, 28, 56 and 180 as determined by OprF/I specific IgG antibody titer measured by ELISA in patients receiving IC43 or placebo
Safety endpoints:
■ Rate of serious adverse events and adverse events during the vaccination period up to 180 days after the first vaccination
■ Systemic tolerability (Vital signs: blood pressure, pulse, body temperature)
■ Local tolerability (local injection site reactions)
■ Safety laboratory parameters (hematology, serum chemistry)
Table A3. System organ class (SOQ: "Hepatobiliary disorders" as defined by MedDRA (v. l l.n and used for the IC43-201 trial.
Hepatobiliary disorders Cholecystitis acute
Hepatobiliary disorders Cholelithiasis
Hepatobiliary disorders Cholestasis
Hepatobiliary disorders Cirrhosis alcoholic
Hepatobiliary disorders Hepatic cirrhosis
Hepatobiliary disorders Hepatic congestion
Hepatobiliary disorders Hepatic failure
Hepatic
Hepatobiliary disorders haemorrhaqe
Hepatobiliary disorders Hepatic steatosis
Hepatobiliary disorders Hepatitis
Hepatobiliary disorders Hepatitis alcoholic
Hepatobiliary disorders Hepatomeqaly
Hepatobiliary disorders Hepatosplenomeqaly
Hepatobiliary disorders Ischaemic hepatitis
Hepatobiliary disorders Jaundice
Hepatobiliary disorders Liver disorder
Hepatobiliary disorders Portal hypertension
Portal vein
Hepatobiliary disorders thrombosis
Hepatobiliary disorders Cytolytic hepatitis
Hepatobiliary disorders Biliary cyst
Hepatobiliary disorders Hepatic ischaemia
Hepatobiliary disorders Biliary dilatation
Hepatobiliary disorders Liver injury
Table A4. System organ class (SOC): Respiratory, thoracic and mediastinal disorders as defined by MedDRA (v. 1 1.1) and used for the IC43-201 trial.
Idiopathic pulmonary fibrosis Diaphragmatic rupture
Laryngeal oedema Acquired diaphragmatic eventration
Laryngospasm Obstructive airways disorder
Lung disorder Increased bronchial secretion
Lung infiltration Thoracic haemorrhage
Pickwickian syndrome Bronchial hyperreactivity
Pleural effusion Organising pneumonia
Pneumonia aspiration Pneumothorax
Pneumonitis
Table A5. System organ class (SOC): "Hepatobiliary disorders" (and others) as defined by MedDRA (v. 18.1) and used for the IC43-202 trial.
SOC: Hepatobiliary disorders (and related disorders)
(according to MedDRA, v. 18.1 ) used for trial IC43-202
HEPATOBILIARY DISORDERS ACUTE HEPATIC FAILURE
HEPATOBILIARY DISORDERS ALCOHOLIC LIVER DISEASE
HEPATOBILIARY DISORDERS AUTOIMMUNE HEPATITIS
HEPATOBILIARY DISORDERS BILE DUCT STONE
HEPATOBILIARY DISORDERS BILIARY CIRRHOSIS PRIMARY
HEPATOBILIARY DISORDERS CHOLANGITIS
HEPATOBILIARY DISORDERS CHOLANGITIS ACUTE
HEPATOBILIARY DISORDERS CHOLANGITIS SCLEROSING
HEPATOBILIARY DISORDERS CHOLECYSTITIS
HEPATOBILIARY DISORDERS CHOLECYSTITIS ACUTE
HEPATOBILIARY DISORDERS CHOLELITHIASIS
HEPATOBILIARY DISORDERS CHOLESTASIS
HEPATOBILIARY DISORDERS CHRONIC HEPATITIS
HEPATOBILIARY DISORDERS CIRRHOSIS ALCOHOLIC
HEPATOBILIARY DISORDERS HEPATIC CIRRHOSIS
HEPATOBILIARY DISORDERS HEPATIC CONGESTION
HEPATOBILIARY DISORDERS HEPATIC CYST
HEPATOBILIARY DISORDERS HEPATIC FAILURE
HEPATOBILIARY DISORDERS HEPATIC FIBROSIS
HEPATOBILIARY DISORDERS HEPATIC FUNCTION ABNORMAL
HEPATOBILIARY DISORDERS HEPATIC NECROSIS
HEPATOBILIARY DISORDERS HEPATIC STEATOSIS
HEPATOBILIARY DISORDERS HEPATITIS
HEPATOBILIARY DISORDERS HEPATITIS ACUTE
HEPATOBILIARY DISORDERS HEPATITIS ALCOHOLIC
HEPATOBILIARY DISORDERS HEPATOCELLULAR INJURY
HEPATOBILIARY DISORDERS HEPATOMEGALY
HEPATOBILIARY DISORDERS HEPATORENAL SYNDROME
HEPATOBILIARY DISORDERS HEPATOSPLENOMEGALY
HEPATOBILIARY DISORDERS HEPATOTOXICITY
HEPATOBILIARY DISORDERS HYPERBILIRUBINAEMIA
HEPATOBILIARY DISORDERS ISCHAEMIC HEPATITIS
HEPATOBILIARY DISORDERS JAUNDICE
HEPATOBILIARY DISORDERS JAUNDICE CHOLESTATIC
HEPATOBILIARY DISORDERS LIVER DISORDER
HEPATOBILIARY DISORDERS PORTAL HYPERTENSION
HEPATOBILIARY DISORDERS PORTAL VEIN THROMBOSIS
HEPATOBILIARY DISORDERS BILIARY CYST
HEPATOBILIARY DISORDERS SUBACUTE HEPATIC FAILURE
HEPATOBILIARY DISORDERS HYDROCHOLECYSTIS
HEPATOBILIARY DISORDERS BILIARY DILATATION
HEPATOBILIARY DISORDERS CHRONIC HEPATIC FAILURE
HEPATOBILIARY DISORDERS GALLBLADDER NECROSIS
HEPATOBILIARY DISORDERS LIVER INJURY
NEOPLASM BILIARY ADENOCARCINOMA
NEOPLASM CHOLANGIOCARCINOMA
NEOPLASM HEPATOCELLULAR CARCINOMA
INVESTIGATIONS HEPATIC ENZYMES INCREASED
INVESTIGATIONS HEPATIC ENZYMES ABNORMAL
INVESTIGATIONS HEPATITIS B VIRUS TEST POSITIVE
INFECTIONS AND INFESTATIONS HEPATITIS A
INFECTIONS AND INFESTATIONS HEPATITIS B
INFECTIONS AND INFESTATIONS HEPATITIS C
INFECTIONS AND INFESTATIONS HEPATITIS VIRAL
INFECTIONS AND INFESTATIONS VIRAL HEPATITIS CARRIER
INFECTIONS AND INFESTATIONS CHRONIC HEPATITIS B
INFECTIONS AND INFESTATIONS PERIHEPATIC ABSCESS
NERVOUS SYSTEM HEPATIC ENCEPHALOPATHY
Table A6. System organ class (SOC): Respiratory, thoracic and mediastinal disorders as defined by MedDRA (v. 18.1) and used for the IC43-202 trial.
PULMONARY ALVEOLAR
BRONCHITIS CHRONIC HAEMORRHAGE
PULMONARY ARTERIAL
BRONCHOPLEURAL FISTULA HYPERTENSION
BRONCHOSPASM PULMONARY CAVITATION
CHEYNE-STO ES RESPIRATION PULMONARY CONGESTION
CHRONIC OBSTRUCTIVE
PULMONARY EMBOLISM PULMONARY DISEASE
CHRONIC RESPIRATORY DISEASE PULMONARY FIBROSIS
CHRONIC RESPIRATORY FAILURE PULMONARY HAEMORRHAGE
COUGH PULMONARY HYPERTENSION
DYSPHONIA PULMONARY MASS
DYSPNOEA PULMONARY NECROSIS
DYSPNOEA EXERTIONAL PULMONARY OEDEMA
DYSPNOEA PAROXYSMAL
RALES NOCTURNAL
EMPHYSEMA RESPIRATORY ACIDOSIS
EPIGLOTTIC OEDEMA RESPIRATORY ALKALOSIS
EPISTAXIS RESPIRATORY ARREST
HAEMOPTYSIS RESPIRATORY DISORDER
HAEMOTHORAX RESPIRATORY DISTRESS
HICCUPS RESPIRATORY FAILURE
HYDROTHORAX RESPIRATORY FATIGUE
RESPIRATORY GAS EXCHANGE
HYPERCAPNIA DISORDER
RESPIRATORY TRACT
HYPERVENTILATION INFLAMMATION
HYPOVENTILATION RESPIRATORY TRACT OEDEMA
HYPOXIA RESTRICTIVE PULMONARY DISEASE
IDIOPATHIC PULMONARY FIBROSIS RHINITIS ALLERGIC
INCREASED VISCOSITY OF
RHONCHI BRONCHIAL SECRETION
INTERSTITIAL LUNG DISEASE SLEEP APNOEA SYNDROME
LARYNGEAL OEDEMA SNEEZING
LOWER RESPIRATORY TRACT
SPUTUM DISCOLOURED INFLAMMATION
LUNG CONSOLIDATION SPUTUM INCREASED
LUNG DISORDER STATUS ASTHMATICUS
LUNG INFILTRATION STRIDOR
LUPUS PNEUMONITIS TACHYPNOEA
MEDIASTINAL SHIFT TONSILLAR HAEMORRHAGE
OBSTRUCTIVE AIRWAYS DISORDER TRACHEOMALACIA
UPPER AIRWAY RESISTANCE
OROPHARYNGEAL PAIN SYNDROME
OROPHARYNGEAL SPASM WHEEZING
ORTHOPNOEA
Example 1 : OprF/I Fusion Protein (1C43) Clinical trial IC43-201
A Phase II placebo-controlled clinical trial (IC43-201) was performed including 399 male and female patients randomized into four cohorts at 33 different medical centers. The patients in the study had been admitted to an ICU with a need for mechanical ventilation for
more than 48 hours and were between 18 and 80 years of age (mean 56.1 years). The subjects were determined by a clinician to have a high probability of survival for at least 48 hours. Criteria used to exclude subjects included a Sequential Organ Failure Assessment (SOFA) index of less than 4 at visit 0; known use of any other investigational or non-registered drug within 30 days prior to administration of the OprF/I vaccine or placebo; subjects having had an organ transplant within the previous 6 months; severe thrombocytopenia or other coagulopathy, which in the opinion of a clinician makes the subject unsuitable for intramuscular injection; pregnancy (positive pregnancy test at visit 0 or visit 1); lactation; subjects having been involuntarily committed to an institution; subjects expecting to receive plasmapheresis or immunoadsorption during the study period; readmission to the ICU during the current total hospital stay on day 0; subjects having been admitted to the ICU within 2 days after surgery; subjects having been admitted to the ICU due to trauma; subjects having elective surgery prior to day 28 following administration of the OprF/I vaccine or placebo. Production and formulation of OprF/I fusion protein (IC43)
The P. aeruginosa OprF/I vaccine (IC43) comprised a fusion protein of the
Pseudomonas outer membrane porin proteins OprF and Oprl. It was expressed as a 224 aa hybrid protein containing an N-terminal His6-tag which is preceded by an alanine and a methionine residue. The N-terminal methionine is cleaved off after expression in E.coli. The primary structure of the expressed fusion protein (excluding the N-terminal methionine) is shown in SEQ ID NO: 3. The OprF/I fusion protein was recombinantly produced in E. coli. Briefly, the OprF/I protein coding sequence was cloned into the multiple cloning site of the expression vector pTRC99A and transformed into E. coli strain XL1. The resulting protein was purified and processed as detailed in WO 2012/126879. The purified OprF/I protein was stored at -80°C in PBS buffer.
Production of vaccine formulations used in clinical trial: The IC43 Drug Product with aluminium hydroxide consisted of the recombinant fusion-protein OprF/I adsorbed to the adjuvant aluminium hydroxide (Alhydrogel 2%). The drug substance, comprising Ala-(His)6- OprFi9o-342-OprI2i-83 protein (OprF/I; SEQ ID NO: 3) in PBS buffer, was thawed overnight at 2-8°C and diluted 1 : 10 with physiological NaCl 0.9% solution to yield a final protein concentration of 100 mcg/mL, followed by 0.2 μιη sterile filtration. Sterile Alhydrogel 2% was added to a final concentration of 400 mcg/mL.
The IC43 Drug Product without aluminium hydroxide was diluted 1 : 10 with NaCl 0.9% solution to yield a final protein concentration of 100 mcg/mL, followed by 0.2 μιη
sterile filtration. The placebo was mixed with NaCl 0.9% solution. To fully mimic the vaccines, the placebo consisted of PBS diluted tenfold with 0.9% Saline, with 400 mcg/ml Al(OH)3 added. The 1 mL dose aliquots of 1.2 mL (extractable volume 1 mL) were aseptically filled into sterile pyrogen-free glass vials. All formulations were stored at 2-8°C before use. The patients in the groups receiving 100 meg IC43 were administered 1 mL of the formulations (with or without alum), whereas those patients receiving 200 meg IC43 were administered 2 mL of the formulation (with alum). Patients in the placebo group received 1 mL of placebo formulation.
Clinical Trial 201
Randomized, placebo-controlled, partially -blinded phase 2 pilot study design: 400 male or female patients admitted to an ICU with need for mechanical ventilation for more than 48 hours, aged between 18 and 80 years were vaccinated intramuscularly on days zero and seven in four treatment groups receiving 100 or 200 meg alum-adjuvanted OprF/I Vaccine (SEQ ID NO: 3), 100 meg non-adjuvanted OprF/I Vaccine (SEQ ID NO: 3) or alum as placebo control (see Table 1, below). Study duration per patient was estimated to be 90 days and overall study duration was estimated to be 12 to 18 months. Primary and secondary endpoints measured were as set forth in Table A2.
Table 1. The number of subjects in the IC43-201 trial and the treatments received.
*These subjects received two doses of the 100 meg IC43 + 400 meg alum formulation.
Table 2. Investigator-confirmed P. aeruginosa infections arising during the course of the IC43-201 trial shown according to those that were identified in the first week, the second week or at any timepoint between the third week until the end of 90 days. Shown are numbers of patients developing invasive infection over the course of the study (including cases of
bacteremia and/or pneumonia) as well as overall infection rates over the course of the study (DS). Overall infection numbers include confirmed bacteremia, pneumonia,
tracheobronchitis, central venous catheter infection, wound infection and urinary tract infection. Also shown are infections present at the start of study (baseline). Some patients had more than one positive P. aeruginosa culture result. In sum, no significant differences in P. aeruginosa infection rates, invasive or total, between patients vaccinated with IC43 or placebo were observed.
DS = during study
Survival analysis for the most relevant subgroups in the IC43-201 trial was performed using the Kaplan-Meier method with the R package "survival". Statistical significance between survival of treated and placebo groups was calculated using the Log-Rank test.
On days zero and seven, the patients were intramuscularly administered a dose of 100 meg non-adjuvanted OprF/I vaccine (SEQ ID NO: 3; 97 patients); 100 meg alum-adjuvanted OprF/I vaccine (SEQ ID NO: 3; 104 patients); 200 meg alum-adjuvanted OprF/I vaccine (SEQ ID NO: 3; 100 patients); or alum only as placebo control (98 patients) (see Table 1).
The study duration per patient was estimated to be 90 days and the overall study duration was estimated to be 12 to 18 months. Patients were evaluated for baseline parameters day 0 (visit 0, V0; see Table 3A), and for during-study parameters on days 7 (VI), 14 (V2), 28 (V3), 42 (V4), 56 (V5), 70 (V6), 90 (V7), and at the time of their ICU discharge (Va) (see Table 3B). Survival at day 28 was selected as a primary endpoint, and survival at day 90 was selected as a secondary endpoint. Subjects were evaluated for Sequential Organ Failure Assessment (SOFA) scores at each of the time points (data not shown).
Prior medication 6 Analgesics, antihypertensives, antibiotics
Physical examination 11 Hepatic, cardiovascular, neurological assessment
Vital signs 1 Body temperature
Laboratory parameters 21 Hemoglobin, hematocrit, fibrinogen
Table 3B: During-study parameters
Variable classification group # variables Examples
Vital signs 1 Body temperature
Laboratory parameters 6 Hemoglobin, fibrinogen, leukocyte count, CRP,
CD4, platelet
ICU 4 Reason, total number of days in ICU
Hospitalization 4 Reason, total number of days in hospital
Ventilation 3 Reason, total number of days with ventilation
Assessment of 2 Avidity index, loglO (OprF/I specific IgG immunogenicity antibodies)
Assessment of efficacy 140 Bacteremia group, bacteria (positive/negative for
P. aeruginosa), SOFA score
Assessment of safety 2 Physical examination change, adverse effects
Concomitant medications 34 Algopyrin, Amikin, Voltaren
Results of IC43-201
The overall survival of patients receiving each of the treatments is shown in Figure 2 and Table 4. The subjects receiving any OprF/I vaccine formulation (with or without alum and at either concentration) had an increased survival (and reduced mortality) throughout the course of evaluation including at the primary and secondary endpoints (days 28 and 90, respectively; see Table 4) as compared to the placebo control group.
Table 4: Overall survival and mortality at days 28 and 90 of subjects receiving the OprF/I vaccine (n = 304) or placebo (n = 98) in the IC43-201 trial. The survival and mortality data for all three groups that received IC43 were combined and compared with placebo-treated subjects.
A hierarchical clustering of subjects was performed to determine whether there were particular parameters, such as parameters related to medical history, laboratory measures, physical examination, or specific immune profiles, or subgroups within the subject population that were associated with enhanced or diminished survival relative to the overall population of subjects or to the population of subjects without the given parameter.
The medical history of each subject participating in the IC43-201 trial was recorded at the start of the trial. In this regard, any underlying disease, related symptoms as well as other pre-existing conditions prior to first vaccination (including any symptoms reported during the first study visit prior to the first vaccination) were considered medical history. In general, the medical history of each subject was reviewed by sites' study staff members at the time of the first study visit and, in this ICU trial, usually based on information provided by the medical record of the patient. Occasionally, site staff became aware of (and reported) single medical history events during the course of the study. Reported medical history was coded by the Contract Research Organization (CRO) Assign using the preferred term (PT) according to the Medical Dictionary for Regulatory Activities (MedDRA). The PT was accordingly allocated to the respective System Organ Class (SOC); e.g., SOC, hepatobiliary disorder or respiratory, thoracic and mediastinal disorders.
As shown in Figure 3 and Table 5, a subset of subjects in the IC43-201 trial who presented with a previous medical history of hepatobiliary disorders and received the OprF/I vaccine (n = 55) had nearly 50% increased survival at day 28 and nearly 40% increased survival at day 90 as compared to placebo-treated subjects with a previous medical history of hepatobiliary disorders (n = 22).
This striking trend of reduced mortality in IC43-treated patients with a history of hepatobiliary disorders was consistent regardless of whether the OprF/I vaccine was administered at a concentration of 100 meg without alum (Figure 4A), at a concentration of 100 meg with alum (Figure 4B), or at a concentration of 200 meg with alum (Figure 4C).
Table 5: Survival and mortality of subsets of patients in the IC43-201 trial with or without a previous medical history of hepatobiliary disorders after receiving the OprF/I vaccine or placebo. Data for all three IC43 treatment groups were combined and compared with placebo-treated subjects in each of the two groups (with and without a history of
IC43 (all) 66% 48% 34% 52%
YES
Placebo 18% 10% 82% 90%
IC43 (all) 77% 52% 23% 48%
NO
Placebo 72% 55% 28% 45%
Subjects presenting at the beginning of the trial with laboratory measures associated with hepatobiliary disorders, such as clinically-relevant (CR) elevations in serum
concentrations of the liver enzymes SGOT and SGPT also showed enhanced survival at both days 28 and 90 after receiving the OprF/I vaccine compared to subjects with elevated SGOT and SGPT which received the placebo (Figures 5A and 5B, respectively). Figure 5C shows the overlap between participating patient groups with clinically relevant (CR) elevations in serum SGOT and SGPT levels and those with a medical history of hepatobiliary disorders. In brief, 24 patients were characterized by all three parameters, whereas 91 patients were characterized by at least one of the parameters associated with disorders in the hepatobiliary system.
Serum levels of SGOT and SGPT were assessed at each study site's local lab. In cases where the test results were not within the normal range specified by the respective lab, the investigator further assessed the result as being either CR or NCR. As an example, abnormal results requiring medical treatment or further diagnostics should be considered CR and thus be documented as an adverse event. In general, normal SGPT and SGOT levels are approximately 10 to 50 units per liter in males and approximately 10 to 35 units per liter serum in female; however, normal ranges differ depending on the protocols and techniques used. During the study, each laboratory routinely provided the normal reference ranges and included them in the report.
Finally, a subset of subjects in the IC43-201 trial which had an abnormal hepatic physical examination at baseline showed enhanced survival at days 28 and 90 after vaccination with OprF/I compared with placebo-treated subjects having with an abnormal hepatic physical examination at baseline (Figure 6A). Figure 6B shows the overlap between participating patient groups with an abnormal hepatic exam and those with a medical history of hepatobiliary disorders. The total number of patients with an abnormal hepatic
examination and/or a previous medical history of hepatobiliary disorders was 1 19, with 46 patients falling into both categories.
Assessment of "normal" or "abnormal" of the physical examination of the liver was done at the discretion of the investigator at each individual site. A hepatic physical
examination in general can include palpation of the liver, estimation of the size of the liver and assessment of the patient for other visible and/or palpable signs and/or symptoms of liver disease.
Shown in Figure 7 are the categories of hepatobiliary disorders identified in the patients taking part in the clinical trial, as well as the number of patients that were affected by the respective disorder.
As shown in Figure 8A, a subset of subjects in the IC43-201 trial who presented with no previous or existing respiratory, thoracic or mediastinal disorders and also received the OprF/I vaccine (n = 120) had nearly 40% increased survival at day 28 and nearly 30% increased survival at day 90 as compared to placebo-treated subjects with no previous or existing RTM disorders (n = 33).
Figure 8B shows the overlap of patients in the IC43-201 trial with no previous or existing respiratory, thoracic or mediastinal disorders (n = 153) and patients having hepatobiliary disorders (n = 77). Only 29 patients met both criteria.
Example 2: OprF/I Fusion Protein (1C43) Clinical trial IC43-202
Here will go the same types of data as outlined in Example 1; such as:
-Numbers of patients
Table x. The number of subjects in the IC43-202 trial and the treatments received.
*These subjects received two doses of the 100 meg IC43 + 400 meg alum formulation.
-Pseudomonas infection data (if available)
Table x. Investigator-confirmed P. aeruginosa infections arising during the course of the IC43-202 trial shown according to those that were identified in the first week, the second
week or at any timepoint between the third week until the end of 90 days. Shown are numbers of patients developing invasive infection over the course of the study (including cases of bacteremia and/or pneumonia) as well as overall infection rates over the course of the study (DS). Overall infection numbers include confirmed bacteremia, pneumonia,
tracheobronchitis, central venous catheter infection, wound infection and urinary tract infection. Also shown are infections present at the start of study (baseline). Some patients had more than one positive P. aeruginosa culture result. In sum, no significant differences in P. aeruginosa infection rates, invasive or total, between patients vaccinated with IC43 or placebo were observed.
* DS = during study
-Survival and mortality (overall)
-Survival and mortality (hepatobiliary disorders)
-Survival (and mortality) (no RTM disorders)
Preferred aspects:
Al . A method of reducing the probability of dying in a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder, comprising administering an effective amount of an OprF antigen to the subject; and/or a method for reducing the mortality of a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder, comprising administering an effective amount of an OprF antigen to the subject.
A2. The OprF antigen for use according to preferred aspect Al, wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
A3. The method according to preferred aspect Al or A2, wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject. A4. The method according to preferred aspects Al to A3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient.
A5. The method according to preferred aspects Al to A4, wherein the subject has an existing Pseudomonas aeruginosa infection or is at risk of acquiring a Pseudomonas aeruginosa infection.
A6. The method according to preferred aspect A5, wherein the existing or acquired Pseudomonas aeruginosa infection is a fulminant Pseudomonas aeruginosa infection. A7. The method according to any one of preferred aspects Al to A6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
A8. The method according to preferred aspect A7, wherein the second administration of the OprF antigen is seven days after the first.
A9. The method according to any one of preferred aspects Al to A8, wherein said subject has been evaluated for said existing hepatobiliary disorder, history of hepatobiliary disorder or indication associated with hepatobiliary disorder, especially wherein said evaluation comprises at least one of the following
-assessment of the medical history of the subject;
-physical examination of the subject;
-measurement of one or more liver proteins in a serum sample from the subject;
-measurement of hepatic excretory function;
-ultrasonography and/or computed tomography scan of the liver;
-angiography; and
-liver biopsy.
A10. The method according to preferred aspect A9, wherein said liver protein is alanine aminotransferase (SGPT), aspartate aminotransferase (SGOT), bilirubin, γ- glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase, and/or albumin.
Al 1. The method according to any one of preferred aspects Al to A 10, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO : 3.
A12. The method according to preferred aspect Al 1, wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3. A13. The method according to preferred aspects Al 1 or A12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200μg/mL OprF antigen.
A14. The method according to any one of preferred aspects Al to A13, further comprising administering an adjuvant, preferably alum.
A15. The method according to any one of preferred aspects Al to A 14, wherein the hepatobiliary disorder is viral hepatitis (A, B or C or other), non-viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, porphyria, Reye's syndrome, sarcoidosis, toxic hepatitis, type
1 glycogen storage disease, tyrosinemia, Wilson disease or a structural or other bile duct disorder.
B 1. The use of an OprF antigen in the manufacture of a pharmaceutical composition for reducing the probability of dying in a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder, comprising administering an effective amount of an OprF antigen to the subject; and/or the use of an OprF antigen in the manufacture of a pharmaceutical composition for reducing the mortality of a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder.
B2. The OprF antigen for use according to preferred aspect B 1, wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
B3. The use according to preferred aspect Bl or B2, wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject.
B4. The use according to any of preferred aspects Bl to B3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient. B5. The use according to any of preferred aspects Bl to B4, wherein the subject has an existing bacterial infection or is at risk of acquiring a bacterial infection.
B6. The use according to preferred aspect B5, wherein the existing or acquired bacterial infection is a Pseudomonas aeruginosa infection.
B7. The use according to any one of preferred aspects B l to B6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
B8. The use according to preferred aspect B7, wherein the second administration of the OprF antigen is seven days after the first.
B9. The use according to any one of preferred aspects B l to B8, wherein said subject has been evaluated for said existing hepatobiliary disorder, history of hepatobiliary disorder or indication associated with hepatobiliary disorder, especially wherein said evaluation comprises at least one of the following
-assessment of the medical history of the subject;
-physical examination of the subject;
-measurement of one or more liver proteins in a serum sample from the subject;
-measurement of hepatic excretory function;
-ultrasonography and/or computed tomography scan of the liver;
-angiography; and
-liver biopsy.
BIO. The use according to preferred aspect B9, wherein said liver protein is alanine aminotransferase (SGPT), aspartate aminotransferase (SGOT), bilirubin, γ- glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase, and/or albumin. B 11. The use according to any one of preferred aspects B 1 to B IO, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
B 12. The use according to preferred aspect B 11 , wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
B 13. The use according to preferred aspects Bl 1 or B 12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200μg/mL OprF antigen.
B14. The use according to any one of preferred aspects B l to B 13, further comprising administering an adjuvant, preferably alum.
B15. The use according to any one of preferred aspects B6 to B 14, wherein the P.
aeruginosa infection is an acute P. aeruginosa infection, wherein the P. aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P. aeruginosa infection.
B 16. The use according to any one of preferred aspects B 1 to B 15, wherein the hepatobiliary disorder is viral hepatitis (A, B or C or other), non-viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, porphyria, Reye's syndrome, sarcoidosis, toxic hepatitis, type 1 glycogen storage disease, tyrosinemia, Wilson disease or a structural or other bile duct disorder.
CI. A pharmaceutical composition comprising an OprF antigen for reducing the probability of dying in a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder, comprising administering an effective amount of an OprF antigen to the subject; and/or the pharmaceutical composition comprising an OprF antigen for reducing the mortality of a subject, wherein the subject has an existing hepatobiliary disorder, a history of a
hepatobiliary disorder or an indication associated with a hepatobiliary disorder.
C2. The pharmaceutical composition according to preferred aspect CI, wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
C3. The pharmaceutical composition according to preferred aspect C2, wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject. C4. The pharmaceutical composition according to any of preferred aspects CI to C3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient.
C5. The pharmaceutical composition according to any of preferred aspects CI to C4, wherein the subject has an existing bacterial infection or is at risk of acquiring a bacterial infection.
C6. The pharmaceutical composition according to preferred aspect C5, wherein the existing or acquired bacterial infection is a Pseudomonas aeruginosa infection. C7. The pharmaceutical composition according to any one of preferred aspects CI to C6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
C8. The pharmaceutical composition according to preferred aspect C7, wherein the second administration of the OprF antigen is seven days after the first.
C9. The pharmaceutical composition according to any one of preferred aspects CI to C8, wherein said subject has been evaluated for said existing hepatobiliary disorder, history of hepatobiliary disorder or indication associated with hepatobiliary disorder, especially wherein said evaluation comprises at least one of the following
-assessment of the medical history of the subject;
-physical examination of the subject;
-measurement of one or more liver proteins in a serum sample from the subject; -measurement of hepatic excretory function;
-ultrasonography and/or computed tomography scan of the liver;
-angiography; and
-liver biopsy.
CIO. The pharmaceutical composition according to preferred aspect C9, wherein said liver protein is alanine aminotransferase (SGPT), aspartate aminotransferase (SGOT), bilirubin, γ- glutamyltransferase, alkaline phosphatase, 5 '-nucleotidase, and/or albumin. CI 1. The pharmaceutical composition according to any one of preferred aspects CI to CIO, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
C12. The pharmaceutical composition according to preferred aspect CI 1, wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
C13. The pharmaceutical composition according to preferred aspects CI 1 or C12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200μg/mL OprF antigen.
C14. The pharmaceutical composition according to any one of preferred aspects CI to C13, further comprising administering an adjuvant, preferably alum.
C15. The pharmaceutical composition according to any one of preferred aspects C6 to CI 4, wherein the P. aeruginosa infection is an acute P. aeruginosa infection, wherein the P.
aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P. aeruginosa infection.
C16. The pharmaceutical composition according to any one of preferred aspects CI to C15, wherein the hepatobiliary disorder is viral hepatitis (A, B or C or other), non-viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis,
cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, porphyria, Reye's syndrome, sarcoidosis, toxic hepatitis, type 1 glycogen storage disease, tyrosinemia, Wilson disease or a structural or other bile duct disorder.
Dl . A method of reducing the probability of dying in a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders, comprising administering an effective amount of an OprF antigen to the subject; and/or a method for reducing the mortality of a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders, comprising administering an effective amount of an OprF antigen to the subject.
D2. The OprF antigen for use according to preferred aspect Dl, wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
D3. The method according to preferred aspect D 1 or D2, wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject.
D4. The method according to preferred aspects Dl or D3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient.
D5. The method according to preferred aspects Dl to D4, wherein the subject has an existing Pseudomonas aeruginosa infection or is at risk of acquiring a Pseudomonas aeruginosa infection. D6. The method according to preferred aspect D5, wherein the existing or acquired Pseudomonas aeruginosa infection is a fulminant Pseudomonas aeruginosa infection.
D7. The method according to any one of preferred aspects Dl to D6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
D8. The method according to preferred aspect D7, wherein the second administration of the OprF antigen is seven days after the first. D9. The method according to any one of preferred aspects Dl to D8, wherein said subject has been evaluated for said existing or medical history of respiratory, thoracic or mediastinal disorders, especially wherein said evaluation comprises at least one of the following
-assessment of the medical history of the subject;
-physical examination of the subject;
-X-ray imaging;
-magnetic resonance imaging (MRI);
-pulse oxiometry;
-computerized axial tomography (CAT) scan;
-electrocardiogram (ECG);
-lung function tests, e.g., spirometry; and
-blood testing.
D10. The method according to preferred aspect D9, wherein said blood testing includes assessing arterial blood gas concentration and/or levels of indicator hormones, e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
Dl 1. The method according to any one of preferred aspects Dl to D10, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3. D12. The method according to preferred aspect D l 1, wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
D13. The method according to preferred aspects Dl 1 or D 12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200μg/mL OprF antigen.
D14. The method according to any one of preferred aspects Dl to D13, further comprising administering an adjuvant, preferably alum. D15. The method according to any one of preferred aspects Dl to D14, wherein the respiratory, thoracic or mediastinal disorder is selected from the following: acquired diaphragmatic eventration, pleural disorder, acute pulmonary edema, pleural effusion, acute respiratory distress syndrome, pleural fibrosis, acute respiratory failure, pleurisy, alveolitis allergic, pleuritic pain, apnea, pneumomediastinum, asphyxia, pneumonia aspiration, aspiration, pneumonitis, asthma, pneumothorax, asthma late onset, productive cough, pleural effusion, atelectasis, prolonged expiration, bronchial disorder, pulmonary alveolar hemorrhage, bronchial fistula, pulmonary arterial hypertension, bronchial hyperreactivity, pulmonary cavitation, bronchial obstruction, pulmonary congestion, bronchiectasis, pulmonary embolism, bronchitis chronic, pulmonary fibrosis, bronchopleural fistula, pulmonary hemorrhage, bronchospasm, pulmonary hypertension, Cheyne-Stokes respiration, pulmonary mass, chronic obstructive pulmonary disease, pulmonary necrosis, chronic respiratory disease, pulmonary edema, chronic respiratory failure, rales, cough, respiratory acidosis, dysphonia, respiratory alkalosis, dyspnea, respiratory arrest, dyspnea exertional, respiratory disorder, dyspnea paroxysmal nocturnal, respiratory distress, emphysema, respiratory failure, epiglottic edema, respiratory fatigue, epistaxis, respiratory gas exchange disorder, hemoptysis, respiratory tract inflammation, haemothorax, respiratory tract edema, hiccups, restrictive pulmonary disease, hydrothorax, rhinitis allergic, hypercapnia, rhonchi, hyperventilation, sleep apnea syndrome, hypoventilation, sneezing, hypoxia, sputum discolored, idiopathic pulmonary fibrosis, sputum increased, increased viscosity of bronchial secretion, status asthmaticus, interstitial lung disease, stridor, laryngeal oedema, tachypnea, lower respiratory tract inflammation, tonsillar hemorrhage, lung consolidation,
tracheomalacia, lung disorder, upper airway resistance syndrome, lung infiltration, wheezing, lupus pneumonitis, mediastinal shift, obstructive airways disorder, oropharyngeal pain, oropharyngeal spasm, orthopnea, pharyngeal hemorrhage, pharyngeal edema, pickwickian syndrome, pleural calcification, laryngospasm, pulmonary microemboli, neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and/or organizing pneumonia.
El . The use of an OprF antigen in the manufacture of a pharmaceutical composition for reducing the probability of dying in a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders, comprising administering an effective amount of an OprF antigen to the subject; and/or the use of an OprF antigen in the manufacture of a pharmaceutical composition for reducing the mortality of a subject, wherein the subject has an no existing or medical history of respiratory, thoracic or mediastinal disorders.
E2. The OprF antigen for use according to preferred aspect El, wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
E3. The use according to preferred aspect El or E2, wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject.
E4. The use according to any of preferred aspects El to E3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient.
E5. The use according to any of preferred aspects El to E4, wherein the subject has an existing bacterial infection or is at risk of acquiring a bacterial infection.
E6. The use according to preferred aspect E5, wherein the existing or acquired bacterial infection is a Pseudomonas aeruginosa infection.
E7. The use according to any one of preferred aspects El to E6, wherein the OprF antigen is administered to the subject more than once, preferably twice. E8. The use according to preferred aspect E7, wherein the second administration of the OprF antigen is seven days after the first.
E9. The use according to any one of preferred aspects El to E8, wherein said subject has been evaluated for an existing or medical history of respiratory, thoracic or mediastinal disorders, especially wherein said evaluation comprises at least one of the following
-assessment of the medical history of the subject;
-physical examination of the subject;
-X-ray imaging;
-magnetic resonance imaging (MRI);
-pulse oxiometry;
-computerized axial tomography (CAT) scan;
-electrocardiogram (ECG);
-lung function tests, e.g., spirometry; and
-blood testing.
E10. The use according to preferred aspect E9, wherein said blood testing includes assessing arterial blood gas concentration and/or levels of indicator hormones, e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
El 1. The use according to any one of preferred aspects El to E10, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
E12. The use according to preferred aspect El 1, wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
E13. The use according to preferred aspects El 1 or E12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200 μg/mL OprF antigen.
E14. The use according to any one of preferred aspects El to E13, further comprising administering an adjuvant, preferably alum.
E15. The use according to any one of preferred aspects E6 to E14, wherein the P.
aeruginosa infection is an acute P. aeruginosa infection, wherein the P. aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P. aeruginosa infection.
E16. The use according to any one of preferred aspects El to E15, wherein the respiratory, thoracic or mediastinal disorder is selected from the following: acquired diaphragmatic eventration, pleural disorder, acute pulmonary edema, pleural effusion, acute respiratory distress syndrome, pleural fibrosis, acute respiratory failure, pleurisy, alveolitis allergic, pleuritic pain, apnea, pneumomediastinum, asphyxia, pneumonia aspiration, aspiration, pneumonitis, asthma, pneumothorax, asthma late onset, productive cough, pleural effusion, atelectasis, prolonged expiration, bronchial disorder, pulmonary alveolar hemorrhage, bronchial fistula, pulmonary arterial hypertension, bronchial hyperreactivity, pulmonary cavitation, bronchial obstruction, pulmonary congestion, bronchiectasis, pulmonary embolism, bronchitis chronic, pulmonary fibrosis, bronchopleural fistula, pulmonary hemorrhage, bronchospasm, pulmonary hypertension, Cheyne-Stokes respiration, pulmonary mass, chronic obstructive pulmonary disease, pulmonary necrosis, chronic respiratory disease, pulmonary edema, chronic respiratory failure, rales, cough, respiratory acidosis, dysphonia, respiratory alkalosis, dyspnea, respiratory arrest, dyspnea exertional, respiratory disorder, dyspnea paroxysmal nocturnal, respiratory distress, emphysema, respiratory failure, epiglottic edema, respiratory fatigue, epistaxis, respiratory gas exchange disorder, hemoptysis, respiratory tract inflammation, haemothorax, respiratory tract edema, hiccups, restrictive pulmonary disease, hydrothorax, rhinitis allergic, hypercapnia, rhonchi, hyperventilation, sleep apnea syndrome, hypoventilation, sneezing, hypoxia, sputum discolored, idiopathic pulmonary fibrosis, sputum increased, increased viscosity of bronchial secretion, status asthmaticus, interstitial lung disease, stridor, laryngeal oedema, tachypnea, lower respiratory tract inflammation, tonsillar hemorrhage, lung consolidation,
tracheomalacia, lung disorder, upper airway resistance syndrome, lung infiltration, wheezing, lupus pneumonitis, mediastinal shift, obstructive airways disorder, oropharyngeal pain, oropharyngeal spasm, orthopnea, pharyngeal hemorrhage, pharyngeal edema, pickwickian syndrome, pleural calcification, laryngospasm, pulmonary microemboli, neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and/or organizing pneumonia.
Fl. A pharmaceutical composition comprising an OprF antigen for reducing the probability of dying in a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders, comprising administering an effective amount of an OprF antigen to the subject; and/or the pharmaceutical composition comprising an OprF antigen for reducing the mortality of a subject, wherein the subject has no existing or medical history of respiratory, thoracic or mediastinal disorders.
F2. The pharmaceutical composition according to preferred aspect Fl, wherein the day 28 mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO).
F3. The pharmaceutical composition according to preferred aspect F2, wherein the reduction of mortality of the subject is due to the treatment or prevention of a Pseudomonas aeruginosa infection in the subject.
F4. The pharmaceutical composition according to any of preferred aspects F l to F3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient. F5. The pharmaceutical composition according to any of preferred aspects F l to F4, wherein the subject has an existing bacterial infection or is at risk of acquiring a bacterial infection.
F6. The pharmaceutical composition according to preferred aspect F5, wherein the existing or acquired bacterial infection is a Pseudomonas aeruginosa infection.
F7. The pharmaceutical composition according to any one of preferred aspects Fl to F6, wherein the OprF antigen is administered to the subject more than once, preferably twice. F8. The pharmaceutical composition according to preferred aspect F7, wherein the second administration of the OprF antigen is seven days after the first.
F9. The pharmaceutical composition according to any one of preferred aspects Fl to F8, wherein said subject has been evaluated for an existing or medical history of respiratory,
thoracic or mediastinal disorders, especially wherein said evaluation comprises at least one of the following
-assessment of the medical history of the subject;
-physical examination of the subject;
-X-ray imaging;
-magnetic resonance imaging (MRI);
-pulse oxiometry;
-computerized axial tomography (CAT) scan;
-electrocardiogram (ECG);
-lung function tests, e.g., spirometry; and
-blood testing.
F10. The pharmaceutical composition according to preferred aspect F9, wherein said blood testing includes assessing arterial blood gas concentration and/or levels of indicator hormones, e.g, atrial natriuretic peptide (ANP) or B-type natriuretic peptide (BNP).
Fl 1. The pharmaceutical composition according to any one of preferred aspects Fl to F10, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEQ ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/I fusion protein set forth as SEQ ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an OprF/I fusion protein comprising at least 80% identity with the OprF/I fusion protein provided by SEQ ID NO: 3.
F12. The pharmaceutical composition according to preferred aspect Fl 1, wherein the amino acid sequence of the OprF/I fusion protein comprises or consists of SEQ ID NO: 3.
F13. The pharmaceutical composition according to preferred aspects Fl 1 or F12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-200μg/mL OprF antigen.
F14. The pharmaceutical composition according to any one of preferred aspects Fl to F13, further comprising administering an adjuvant, preferably alum.
F15. The pharmaceutical composition according to any one of preferred aspects F6 to F14, wherein the P. aeruginosa infection is an acute P. aeruginosa infection, wherein the P. aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P. aeruginosa infection.
F16. The pharmaceutical composition according to any one of preferred aspects Fl to F15, wherein the respiratory, thoracic or mediastinal disorder is selected from the following: acquired diaphragmatic eventration, pleural disorder, acute pulmonary edema, pleural effusion, acute respiratory distress syndrome, pleural fibrosis, acute respiratory failure, pleurisy, alveolitis allergic, pleuritic pain, apnea, pneumomediastinum, asphyxia, pneumonia aspiration, aspiration, pneumonitis, asthma, pneumothorax, asthma late onset, productive cough, pleural effusion, atelectasis, prolonged expiration, bronchial disorder, pulmonary alveolar hemorrhage, bronchial fistula, pulmonary arterial hypertension, bronchial hyperreactivity, pulmonary cavitation, bronchial obstruction, pulmonary congestion, bronchiectasis, pulmonary embolism, bronchitis chronic, pulmonary fibrosis, bronchopleural fistula, pulmonary hemorrhage, bronchospasm, pulmonary hypertension, Cheyne-Stokes respiration, pulmonary mass, chronic obstructive pulmonary disease, pulmonary necrosis, chronic respiratory disease, pulmonary edema, chronic respiratory failure, rales, cough, respiratory acidosis, dysphonia, respiratory alkalosis, dyspnea, respiratory arrest, dyspnea exertional, respiratory disorder, dyspnea paroxysmal nocturnal, respiratory distress, emphysema, respiratory failure, epiglottic edema, respiratory fatigue, epistaxis, respiratory gas exchange disorder, hemoptysis, respiratory tract inflammation, haemothorax, respiratory tract edema, hiccups, restrictive pulmonary disease, hydrothorax, rhinitis allergic, hypercapnia, rhonchi, hyperventilation, sleep apnea syndrome, hypoventilation, sneezing, hypoxia, sputum discolored, idiopathic pulmonary fibrosis, sputum increased, increased viscosity of bronchial secretion, status asthmaticus, interstitial lung disease, stridor, laryngeal oedema, tachypnea, lower respiratory tract inflammation, tonsillar hemorrhage, lung consolidation, tracheomalacia, lung disorder, upper airway resistance syndrome, lung infiltration, wheezing, lupus pneumonitis, mediastinal shift, obstructive airways disorder, oropharyngeal pain, oropharyngeal spasm, orthopnea, pharyngeal hemorrhage, pharyngeal edema, pickwickian syndrome, pleural calcification, laryngospasm, pulmonary microemboli, neonatal hypoxia, tracheal stenosis, pulmonary hematoma, diaphragmatic rupture, increased bronchial secretion, thoracic hemorrhage and/or organizing pneumonia.
Claims
1. An OprF antigen for use in the reduction of the probability of dying in a subject, wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder.
2. An OprF antigen for use in the reduction of mortality of a subject, wherein the day 28 reduced mortality of the subject is lower than 95, and wherein mortality is defined as the ratio of the mortality in treated subjects versus the mortality in placebo controlled subjects times one hundred (xlOO), and wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder.
3. The OprF antigen for use in the prevention and/or treatment of a Pseudomonas aeruginosa infection in a subject, and wherein the subject has an existing hepatobiliary disorder, a history of a hepatobiliary disorder or an indication associated with a hepatobiliary disorder..
4. The OprF antigen for use according to any of claims 1 to 3, wherein the subject is a hospitalized patient, an ICU patient or a ventilated ICU patient.
5. The OprF antigen for use according to claims 1 to 4, wherein the subject has an existing Pseudomonas aeruginosa infection or is at risk of acquiring a Pseudomonas aeruginosa infection.
6. The OprF antigen for use according to claim 5, wherein the existing or acquired
Pseudomonas aeruginosa infection is a fulminant Pseudomonas aeruginosa infection.
7. The OprF antigen for use according to any one of claims 1 to 6, wherein the OprF antigen is administered to the subject more than once, preferably twice.
8. The OprF antigen for use according to claim 7, wherein the second administration of the OprF antigen is seven days after the first.
9. The OprF antigen for use according to any one of claims 1 to 8, wherein said subject has been evaluated for said existing hepatobiliary disorder, history of hepatobiliary disorder or indication associated with hepatobiliary disorder, especially wherein said evaluation comprises at least one of the following
-assessment of the medical history of the subject;
-physical examination of the subject;
-measurement of one or more liver proteins in a serum sample from the subject;
-measurement of hepatic excretory function;
-ultrasonography and/or computed tomography scan of the liver;
-angiography; and
-liver biopsy.
10. The OprF antigen for use according to claim 9, wherein said liver protein is alanine aminotransferase (SGPT), aspartate aminotransferase (SGOT), bilirubin, γ-glutamyltransferase, alkaline phosphatase, 5'-nucleotidase, and/or albumin.
11. The OprF antigen for use according to any one of claims 1 to 10, wherein the OprF antigen is selected from the group consisting of the OprF polypeptide set forth as SEO ID NO: 1 or an antigenic fragment thereof; the OprF/l fusion protein set forth as SEO ID NO: 3 or an antigenic fragment thereof; a nucleic acid encoding the OprF polypeptide set forth as SEO ID NO: 1 or an antigenic fragment thereof; a nucleic acid encoding the OprF/l fusion protein set forth as SEO ID NO: 3 or an antigenic fragment thereof; and an immunogenic variant of an
OprF/l fusion protein comprising at least 80% identity with the OprF/l fusion protein provided by SEQ. ID NO : 3.
12. The OprF antigen for use according to claim 11, wherein the amino acid sequence of the OprF/l fusion protein comprises or consists of SEO ID NO : 3.
13. The OprF antigen for use according to claim 11 or 12, wherein the OprF antigen is administered in a pharmaceutical composition comprising 100-20C^g/m L OprF antigen.
14. The OprF antigen for use according to any one of claims 1 to 13, further comprising administering an adjuvant, preferably alum.
15. The OprF antigen for use according to any one of claims 6 to 14, wherein the P.
aeruginosa infection is an acute P. aeruginosa infection, wherein the P. aeruginosa infection is a fulminant P. aeruginosa infection or wherein the P. aeruginosa infection is a chronic P.
aeruginosa infection.
16. The OprF antigen for use according to any one of claims 1 to 15, wherein the
hepatobiliary disorder is viral hepatitis (A, B or C or other), non-viral hepatitis, liver transplant, cirrhosis, chronic liver disease, a metabolic liver disorder, a vascular liver disorder, acute toxic liver injury, ischemic liver injury, cholestasis, jaundice, cholecystitis, cholelithiasis, hepatic failure, hepatic fibrosis, bile duct infection, cancer of the liver, cancer of the biliary tract, Alagille syndrome, alpha 1 anti-trypsin deficiency, autoimmune hepatitis, cystic disease of the liver, fatty liver disease, galactosemia, gallstones, Gilbert's syndrome, hemochromatosis, liver disease in pregnancy, neonatal hepatitis, primary biliary cirrhosis, primary sclerosing cholangitis, porphyria, Reye's syndrome, sarcoidosis, toxic hepatitis, type 1 glycogen storage disease, tyrosinemia, Wilson disease or a structural or other bile duct disorder.
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP15170486 | 2015-06-03 | ||
| EP15170486.3 | 2015-06-03 | ||
| EP15195697 | 2015-11-20 | ||
| EP15195697.6 | 2015-11-20 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2016193402A1 true WO2016193402A1 (en) | 2016-12-08 |
Family
ID=56121053
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2016/062574 Ceased WO2016193402A1 (en) | 2015-06-03 | 2016-06-02 | Pseudomonas vaccine |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2016193402A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN117940554A (en) * | 2022-06-30 | 2024-04-26 | 上海羽冠生物技术有限公司 | Live bacterial strains of the genus Pseudomonas |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0717106A1 (en) * | 1994-12-16 | 1996-06-19 | BEHRINGWERKE Aktiengesellschaft | Immunogenic hybrid protein OprF-Oprl derived from Pseudomonas aeruginosa membrane proteins |
| WO2012084272A1 (en) | 2010-12-23 | 2012-06-28 | Intercell Ag | Oprf/i agents and their use in hospitalized and other patients |
| WO2012126879A1 (en) | 2011-03-18 | 2012-09-27 | Intercell Ag | Novel oprf/i fusion proteins, their preparation and use |
-
2016
- 2016-06-02 WO PCT/EP2016/062574 patent/WO2016193402A1/en not_active Ceased
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EP0717106A1 (en) * | 1994-12-16 | 1996-06-19 | BEHRINGWERKE Aktiengesellschaft | Immunogenic hybrid protein OprF-Oprl derived from Pseudomonas aeruginosa membrane proteins |
| WO2012084272A1 (en) | 2010-12-23 | 2012-06-28 | Intercell Ag | Oprf/i agents and their use in hospitalized and other patients |
| WO2012126879A1 (en) | 2011-03-18 | 2012-09-27 | Intercell Ag | Novel oprf/i fusion proteins, their preparation and use |
Non-Patent Citations (19)
| Title |
|---|
| "Comparison of biosequences", ADV. APPL. MATH., vol. 2, 1981, pages 482 - 489 |
| "Remington: The Science and Practice of Pharmacy. 20th ed.", 2000, MACK PUBLISHING CO. |
| ALTSCHUL ET AL.: "Basic local alignment search tool", J. MOL. BIOL., vol. 215, 1990, pages 403 - 410, XP002949123, DOI: doi:10.1006/jmbi.1990.9999 |
| ALTSCHUL ET AL.: "Gapped BLAST and PSI-BLAST: a new generation of protein database search programs", NUC. ACIDS RES., vol. 25, 1997, pages 3389 - 3402, XP002905950, DOI: doi:10.1093/nar/25.17.3389 |
| ANONYMOUS: "Virulence", WIKIPEDIA: THE FREE ENCYCLOPEDIA., 7 March 2015 (2015-03-07) |
| BAUMANN U ET AL: "Recombinant OprF-OprI as a vaccine against Pseudomonas aeruginosa infections", VACCINE, ELSEVIER LTD, GB, vol. 22, no. 7, 17 February 2004 (2004-02-17), pages 840 - 847, XP004487437, ISSN: 0264-410X, DOI: 10.1016/J.VACCINE.2003.11.029 * |
| BRENT ET AL.: "Current Protocols in Molecular Biology", 2003, JOHN WILEY & SONS, INC. |
| BUMANN D ET AL: "Systemic, nasal and oral live vaccines against Pseudomonas aeruginosa: A clinical trial of immunogenicity in lower airways of human volunteers", VACCINE, ELSEVIER LTD, GB, vol. 28, no. 3, 8 January 2010 (2010-01-08), pages 707 - 713, XP026793740, ISSN: 0264-410X, [retrieved on 20091101] * |
| CHASTRE; FAGON: "Ventilator-associated pneumonia", RESPIR. CRIT. CARE MED., vol. 165, 2002, pages 867 - 903 |
| CHEVALUPPA ET AL., INTERNATIONAL JOURNAL OF INFECTIOUS DISEASES, vol. 14, 2010, pages E857 - E867 |
| DING, B. ET AL.: "OprF/I vaccinated sera inhibit binding of human interferon-gamma to Pseudomonas aeruginosa", VACCINE, vol. 28, 2010, pages 4119 - 4122, XP055192691, DOI: doi:10.1016/j.vaccine.2010.04.028 |
| FOREMAN ET AL.: "Cirrhosis as a risk factor for sepsis and death", CHEST, vol. 124, 2003, pages 1016 - 1020 |
| MANSOURI ET AL: "Safety and immunogenicity of a Pseudomonas aeruginosa hybrid outer membrane protein F-I vaccine in human volunteers.", INFECTION AND IMMUNITY, vol. 67, no. 3, 1 March 1999 (1999-03-01), pages 1461 - 70, XP055002322, ISSN: 0019-9567 * |
| NEEDLEMAN; WUNSCH: "A general method applicable to the search for similarities in the amino acid sequence of two proteins", J. MOL. BIOL., vol. 48, 1970, pages 443 - 53, XP024011703, DOI: doi:10.1016/0022-2836(70)90057-4 |
| PEARSON; LIPMAN: "Improved tools for biological sequence comparison", PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 2444 - 8, XP002060460, DOI: doi:10.1073/pnas.85.8.2444 |
| SHELLMAN ET AL.: "Prognosis of patients with cirrhosis and chronic liver disease admitted to the medical intensive care unit", CRIT. CARE MED., vol. 16, 1988, pages 671 - 678 |
| VON SPECHT B U ET AL: "Immunogenic efficacy of differently produced recombinant vaccines candidates against Pseudomonas aeruginosa infections", JOURNAL OF BIOTECHNOLOGY, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 83, no. 1-2, 29 September 2000 (2000-09-29), pages 3 - 12, XP004212145, ISSN: 0168-1656, DOI: 10.1016/S0168-1656(00)00311-4 * |
| WU ET AL.: "Recognition of Host Immune Activation by Pseudomonas aeruginosa", SCIENCE, vol. 309, 2005, pages 774 - 777 |
| WUNSCH ET AL.: "The epidemiology of mechanical ventilation use in the United States", CRIT. CARE MED, vol. 38, 2010, pages 1947 - 1953 |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN117940554A (en) * | 2022-06-30 | 2024-04-26 | 上海羽冠生物技术有限公司 | Live bacterial strains of the genus Pseudomonas |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| EP4410843A1 (en) | Recombinant fusion protein derived from hr region of s2 protein of sars-cov-2 and application of recombinant fusion protein | |
| JP2025107346A (en) | Use of thymosin alpha for the treatment of sepsis - Patent Application 20070233633 | |
| JP2020515587A (en) | Immunogenic compositions, uses and methods of treatment | |
| JP6243367B2 (en) | Non-typeable Haemophilus influenzae vaccine and uses thereof | |
| WO2016193402A1 (en) | Pseudomonas vaccine | |
| Sorichter et al. | Immune responses in the airways by nasal vaccination with systemic boosting against Pseudomonas aeruginosa in chronic lung disease | |
| RU2593953C2 (en) | Vaccine with live bacterial isolates for systemic administration | |
| KR20220047332A (en) | Therapeutic effect and its administration route | |
| JP2010501599A (en) | Vaccines and methods for the treatment or prevention of Salmonella bacterial infections in vertebrate subjects | |
| CN110891597B (en) | Composition for preventing and treating poultry synovium infection | |
| CN113046384A (en) | Construction method of broad-spectrum antiviral recombinant salmonella | |
| JP2017536823A (en) | Meningitis B vaccine | |
| US20240299528A1 (en) | A dna plasmid sars-corona virus-2/covid-19 vaccine | |
| JP5520389B2 (en) | Route of administration for animal protection composition against Rhodococcus equi | |
| JP2017509597A (en) | Tolerogenic compositions and uses thereof | |
| US20220204570A1 (en) | Cra4s1 gene, encoded cra4s1 protein, and application | |
| EP2501396A1 (en) | LOW ANTIGEN-DOSE IMMUNIZATION FOR MAXIMIZING T-HELPER CELL 1 (T h1) IMMUNITY AGAINST DISEASE | |
| TWI850940B (en) | Recombinant lactic acid bacteria and their use for simultaneous treatment and/or prevention of dust mite and cockroach allergy | |
| US20140023684A1 (en) | Vaccine Against Pasteurellaceae | |
| JP6867623B2 (en) | Whooping cough model animal and its preparation method, and method using whooping cough model animal | |
| EP2675272A1 (en) | Thymosin alpha peptide for preventing, reducing the severity of, and treating infection | |
| Hashimoto et al. | Hypertonic intranasal vaccines gain nasal epithelia access to exert strong immunogenicity | |
| US12502428B1 (en) | Immunotherapeutic composition for prevention of obesity, nonalcoholic fatty liver disease and hypertriglyceridemia, and methods of use and preparation thereof | |
| KR20240126800A (en) | COVID-19 Vaccine | |
| JP2009533478A (en) | Use of the TRAP protein itself as an active ingredient for the manufacture of a medicament for the treatment of Staphylococcus aureus infections |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 16728885 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 16728885 Country of ref document: EP Kind code of ref document: A1 |