[go: up one dir, main page]

WO2025226628A1 - Use of il-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject - Google Patents

Use of il-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject

Info

Publication number
WO2025226628A1
WO2025226628A1 PCT/US2025/025686 US2025025686W WO2025226628A1 WO 2025226628 A1 WO2025226628 A1 WO 2025226628A1 US 2025025686 W US2025025686 W US 2025025686W WO 2025226628 A1 WO2025226628 A1 WO 2025226628A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
siltuximab
fragment
treatment
dose
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.)
Pending
Application number
PCT/US2025/025686
Other languages
French (fr)
Inventor
Derek E. BYERS
Ramsey HACHEM
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Recordati Rare Diseases Inc
Original Assignee
Recordati Rare Diseases Inc
Priority date (The priority date 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 date listed.)
Filing date
Publication date
Application filed by Recordati Rare Diseases Inc filed Critical Recordati Rare Diseases Inc
Publication of WO2025226628A1 publication Critical patent/WO2025226628A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/244Interleukins [IL]
    • C07K16/248IL-6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • This invention relates to the use of IL-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject.
  • this invention relates to methods of treatment of antibody mediated rejection (AMR) of a lung transplant in a subject in need thereof comprising a combination therapy of (i) administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6; and (ii) administration to the subject of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
  • AMR antibody mediated rejection
  • Lung transplantation is the ultimate treatment option for individuals with advanced lung disease, and approximately 2,500 people undergo lung transplantation annually in the US (1). LT improves quality of life and survival; however, long-term outcomes remain disappointing, and the median survival after LT is 6.7 years (2).
  • Chronic lung allograft dysfunction is the leading cause of death beyond the first year after LT (2,3).
  • BOS bronchiolitis obliterans syndrome
  • RAS restrictive allograft syndrome
  • a mixed phenotype with features of both BOS and RAS is also recognized.
  • CLAD typically follows a progressive clinical course that culminates in respiratory failure and death or retransplantation.
  • CLAD chronic lung disease
  • GEF gastroesophageal reflux disease
  • PWD primary graft dysfunction
  • ACR acute cellular rejection
  • DSA donor-specific antibodies
  • HLA human leukocyte antigens
  • AMR antibody-mediated rejection
  • CARV community-acquired respiratory viral
  • AMR is an increasingly recognized form of lung rejection, in large part because of improved awareness and the development of a standardized definition that facilitates research and communication between centres.
  • ISHLT International Society for Heart and Lung Transplantation
  • DSA abnormal lung histology
  • allograft dysfunction complement component 4d (C4d) deposition
  • C4d complement component 4d
  • Diagnostic certainty depends on the number of criteria present; for example, a diagnosis of definite AMR is made if all 5 criteria are present, and a diagnosis of probable AMR is made if 4 of the 5 criteria are present.
  • a variety of immunosuppressive treatments focused on antibody-depletion have been used, and generally, the standard of care for AMR patients, is a multi-drug regimen including various combinations of high-dose corticosteroids, intravenous immune globulin (IVIG), Rituximab, Bortezomib or Carfilzomib, anti-thymocyte globulin (ATG), and plasma exchange (PLEX) (20, 21, 24-28).
  • IVIG intravenous immune globulin
  • Rituximab Rituximab
  • Bortezomib or Carfilzomib anti-thymocyte globulin
  • PLEX plasma exchange
  • outcomes after the diagnosis of AMR are dismal. Approximately 20% of patients die within 30 days, 50% die within 1 year, and 80% die within 2 years of the diagnosis of AMR (20, 21, 27).
  • An object of the invention is to provide a treatment for antibody mediated rejection (AMR) in lung transplant recipients.
  • AMR antibody mediated rejection
  • the present invention provides a method of treatment of antibody mediated rejection (AMR) of a lung transplant, the method comprising a combination therapy of: (i) administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6; and (ii) administration to the subject of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
  • AMR antibody mediated rejection
  • the present invention provides an anti-IL-6 antibody or fragment for use in treatment of an antibody mediated rejection (AMR) of a lung transplant, wherein the treatment comprises a combination therapy of administration of the antibody or fragment and administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
  • AMR antibody mediated rejection
  • the present invention provides a use of an anti-IL-6 antibody or fragment in treatment of antibody mediated rejection (AMR) of a lung transplant, wherein the treatment comprises a combination therapy of administration of the antibody or fragment and administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
  • AMR antibody mediated rejection
  • the present invention provides a use of an anti-IL-6 antibody or fragment for the manufacture of a medicament for treatment of antibody mediated rejection (AMR) of a lung transplant, wherein the medicament is to be administered in a combination therapy with administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
  • AMR antibody mediated rejection
  • IL-6 is a pleiotropic cytokine that can be secreted by many cell types upon appropriate stimulation and promotes inflammatory, immune, and fibrotic responses. Different modes of IL-6 signalling exist.
  • IL-6 binds to its receptor (IL-6R), which is devoid of signalling capacity, and the complex of IL-6 and IL6R binds to glycoprotein 130 (gpl30) which dimerizes and initiates intracellular signalling (29).
  • IL-6R its receptor
  • gpl30 glycoprotein 130
  • sIL-6R soluble form of IL-6R
  • gpl30 inducing dimerization and intracellular signalling
  • Trans-presentation is a third mode of IL-6 signalling where IL-6 binds to IL-6R in the cytoplasm of dendritic cells and the complex is transported to the plasma membrane where it binds to gpl30 expressed by CD4 + T-cells (31). Trans-presentation was recently discovered in mice but has not been confirmed in humans.
  • IL-6 expands effector and memory CD8 + T-cells by augmenting the expression of IL-2 and receptor CD122/CD25 and is essential for the differentiation of naive CD4 + T-cells towards the Thl7 phenotype (32-34).
  • IL-17 produced by Thl7 cells promotes neutrophil proliferation and migration, endothelial cell activation, and fibroblast activation and proliferation which result in cytopathic responses implicated in acute and chronic rejection (33, 34).
  • IL-6 was initially identified as B-cell stimulating factor 2 (BSF-2) because of its role in promoting antibody synthesis by B-cells (35-37).
  • IL-6 in conjunction with other cytokines is responsible for normal antibody production and is critical for the induction of follicular helper T-cells (Tfh) as well as the production of IL-21 which regulates immunoglobulin synthesis (38, 39). IL- 6 is also crucial for B-cell differentiation into plasmablasts and for enhancing plasmablast survival (40, 41).
  • Blocking IL-6 signalling effectively reduces B-cell activation, plasmablast differentiation, and antibody production (42-44).
  • IL-6 also promotes innate immune responses.
  • NK cells express IL-6R and are activated by IL-6 to induce endothelial cell cytotoxicity (45).
  • IL-6 promotes fibrosis by inducing the differentiation of fibroblasts into myofibroblasts and participating in vascular smooth muscle cell and endothelial cell proliferation and activation (46). These are all important components of the pathogenesis of AMR.
  • High concentrations of IL-6 in bronchoalveolar lavage (BAL) fluid and peripheral blood are seen with severe PGD after LT and are associated with prolonged length of stay in the intensive care unit and the hospital.
  • IL-6 levels have not been consistently associated with acute cellular rejection and there have been no published results of IL-6 levels in AMR after LT.
  • the inventors have surprisingly found that blocking IL-6 signalling may be a potential therapeutic approach for treatment of AMR in LT patients, especially when delivered with the standard of care for AMR in LT patients.
  • IL-6 signalling is a therapeutic target in solid organ transplantation.
  • NEP non-human primate
  • MMF mycophenolate mofetil
  • steroids and all had DSA at the time of rejection (47).
  • Tocilizumab IL6R inhibitor
  • kidney 44, 49-54)
  • cardiac 48
  • Clazakizumab (IL6 inhibitor) has been studied for AMR in kidney transplants (55, 56).
  • RCT pilot randomised controlled trial
  • clazakizumab attenuated the decline in GFR and depleted DSA; however, 25% of treated patients developed serious infections and 10% developed complicated diverticulitis (56).
  • a Phase 3 RCT examining the efficacy and safety of clazakizumab for the treatment of chronic AMR after kidney transplantation is currently enrolling patients (NCT03744910).
  • the inventors have surprisingly found that from immunofluorescence staining for IL-6 in lung biopsies of patients with AMR, there is evidence of intrapulmonary IL-6 expression. These data illustrate increased IL-6 levels in serum and increased expression in lung allografts with AMR.
  • IL-6 inhibitors available today for treatment of rheumatoid arthritis, giant cell arteritis, systemic sclerosis-associated interstitial lung disease, cytokine release syndrome, idiopathic multicentric Castleman's disease (MCD), and neuromyelitis optica.
  • Siltuximab which is the only monoclonal antibody to IL-6 that is FDA-approved. In previous studies, Siltuximab has been used in patients with hematologic diseases/malignancies, solid tumors and MCD (Multicentric Castleman's Disease).
  • Siltuximab is a chimeric (human-murine) immunoglobulin G monoclonal antibody that binds IL-6 and is approved for the treatment of idiopathic MCD. Siltuximab prevents IL-6 from binding to both soluble and membrane bound IL-6R with high affinity and specificity resulting in deactivation of IL-6.
  • Siltuximab has been no published reports describing the use of Siltuximab after LT or for the treatment of AMR after any solid organ transplant. Further, there have been no published reports of use of Siltuximab in combination with routine treatment for AMR.
  • IL-6 inhibitors such as but not limited to Siltuximab
  • Siltuximab IL-6 inhibitors
  • These IL-6 inhibitors, such as Siltuximab, are to be used in combination with the routine immunosuppressive therapy (standard of care) for AMR in lung transplant recipients.
  • any method, use or treatment described herein can be implemented with respect to any other method, use or treatment described herein. All features disclosed herein in connection with any particular aspect are applicable to each of the other aspects, mutatis mutandis. In particular, all features disclosed herein in connection with the first aspect are applicable to each of the second to fourth aspects, mutatis mutandis. Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description.
  • the first aspect of the invention is a method of treatment of antibody mediated rejection (AMR) of a lung transplant in a subject in need thereof, the method comprising a combination therapy of:
  • treatment refers to therapeutic (curative) treatment including amelioration. Treatment also includes stopping the disease from developing or slowing further progression of the disease. For example, treatment may include preventing symptoms from worsening.
  • prevention refers to prophylaxis treatment i.e., action taken to prevent disease.
  • a patient a subject or a recipient
  • a patient a human patient, subject or recipient.
  • the patient is an adult i.e. > 18 years old at the commencement of the treatment.
  • the patient may be a paediatric patient i.e. ⁇ 18 years old at the commencement of the treatment.
  • the subject is a human.
  • “combination therapy” means that both (i) an antibody or fragment which is capable of inhibiting human IL-6; and (ii) an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6, are administered in combination as a treatment to the patient.
  • an immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6, are administered in combination as a treatment to the patient.
  • AMR antibody-mediated rejection
  • ISHLT International Society for Heart and Lung Transplantation
  • a process of immune activation whereby allospecific B-cells and plasma cells produce antibodies directed against donor lung antigens, is central to the concept of pulmonary AMR.
  • the antigen-antibody complex may result in an amplified immune response, via both complement-dependent and independent pathways, which may result in lung tissue pathology and graft dysfunction to a variable degree.
  • Complement is a multifunctional system of receptors, regulators and effector molecules that may amplify both innate and adaptive immunity contributing to the pathogenesis of AMR.
  • Pulmonary AMR may be clinical (allograft dysfunction which can be asymptomatic) or sub-clinical (normal allograft function). Both clinical and sub-clinical AMR were further sub-categorized into 3 mutually exclusive possibilities (definite, probable, and possible) by the ISHLT. These categories are based on the degree of certainty related to the presence or absence of a number of pathologic, serologic, clinical and immunologic criteria and are summarised in Table 1 (clinical) and Table 2 (subclinical).
  • Table 1 Definition and Diagnostic Certainty of Clinical Pulmonary Antibody-mediated Rejection (23); + means item present; - means item absent or missing.
  • Table 2 Definition and Diagnostic Certainty of Sub-clinical Pulmonary Antibody- mediated Rejection (23); + means item present; - means item absent or missing.
  • DSA Donor-specific antibodies
  • a diagnosis of definite clinical pulmonary AMR may be made if all five criteria are present, and a diagnosis of probable clinical pulmonary AMR may be made if any four of the five criteria are present.
  • a diagnosis of definite sub-clinical pulmonary AMR may be made if all three criteria (abnormal lung histology, lung biopsy C4d, and donorspecific antibodies) are present, and a diagnosis of probable sub-clinical pulmonary AMR may be made if any two of the three criteria (abnormal lung histology, lung biopsy C4d, and donor-specific antibodies) are present. Measurement and determination of each of these criteria can be achieved by any appropriate method known in the art.
  • the AMR is probable AMR or definite AMR.
  • AMR is increasingly recognized as a potential contributing factor to acute lung allograft dysfunction and the development of CLAD in pediatric lung transplant recipients (> 18 years of age). Although the frequency is unknown in children, AMR has clearly been documented across all pediatric age groups from infancy to early adulthood. Adult diagnostic criteria for lung allograft rejection are often applied to pediatric patients, with recent confirmation that these criteria are consistent in children.
  • pulmonary AMR may stabilize, and/or improve. Improvement may be partial or complete. A complete response may be a return to baseline graft function if applicable, abolition of DSA titers and reversal of pathologic changes. A partial response may be an improvement in graft function if applicable, but not all parameters return to baseline. Stabilization may be defined as no further clinical deterioration.
  • HLA Donor-specific anti-human leukocyte antigen
  • AMR antibody-mediated rejection
  • donor-specific antibodies is intended to mean antibodies to donor human leukocyte antigens (HLA) with a Mean Fluorescence Intensity > 1000. These can be either pre-existing or de novo. These antibodies can be measured by any appropriate method known in the art.
  • HLA human leukocyte antigens
  • the AMR comprises the presence of donor-specific antibodies to human leukocyte antigens in the subject and/or wherein the AMR comprises allograft dysfunction.
  • allograft dysfunction may, for example, be considered as chronic lung allograft dysfunction.
  • allograft survival may be defined as freedom of the transplant recipient from death or retransplantation.
  • the administration of the combination therapy described herein reduces one or more of the severity of allograft dysfunction, the severity of abnormal lung histology, and the amount of donor-specific antibodies in the subject.
  • the administration of the combination therapy described herein reduces the severity of allograft dysfunction and the amount of donorspecific antibodies in the subject, and optionally reduces the severity of abnormal lung histology.
  • the administration of the combination therapy described herein may reduce or eliminate one, two, three, four or all five criteria for AMR, such that the subject would no longer have definite AMR or probable AMR.
  • lung transplant we mean lung transplantation, or pulmonary transplantation, which is a surgical procedure in which one (single lung transplant) or both (bilateral lung transplant) lungs are replaced by lungs from a donor.
  • Donor lungs can be retrieved from a living or deceased donor, such as a donor that has been declared brain dead. A living donor can only donate one lung lobe.
  • lobe transplant Another type of lung transplant that is relevant to the present invention is a lobe transplant, which is a surgery in which part of a living or deceased donor's lung is removed and used to replace the recipient's diseased lung.
  • living donation this procedure requires the donation of lobes from two different people, replacing a lung on each side of the recipient.
  • Donors who have been properly screened should be able to maintain a normal quality of life despite the reduction in lung volume.
  • deceased lobar transplantation one donor can provide both lobes.
  • a donor-recipient matching system may be used to determine an appropriate donor for a patient.
  • the system administered by the United Network for Organ Sharing finds an appropriate match based on specific criteria, including blood type, size of organ compared with chest cavity, geographic distance between donor organ and transplant recipient, severity of the recipient's lung disease, recipient's overall health, and likelihood that the transplant will be successful.
  • the method is initiated after the transplant of a lung from a donor.
  • the lung transplant may be a single or bilateral lung transplant.
  • a bilateral lung transplant (BLT) is sometimes referred to as a double lung transplant or a sequential single lung transplant.
  • Lung transplantation may be used as a therapeutic measure of last resort for patients with end-stage lung disease who have exhausted all other available treatments without improvement.
  • a variety of conditions may make such surgery necessary.
  • COPD chronic obstructive pulmonary disease
  • emphysema including emphysema; idiopathic pulmonary fibrosis; cystic fibrosis; idiopathic (formerly known as "primary") pulmonary hypertension; alpha 1-antitrypsin deficiency; replacing previously transplanted lungs that have since failed; and other causes, including bronchiectasis and sarcoidosis.
  • a double lung transplant may be required for example in patients with cystic fibrosis.
  • Some patients may require a combined heart and lung transplantation, where the patient receives a heart and two lungs. This type of transplant may be required for example where a patient has a congenital condition(s) or patients in whom a lung condition has caused significant heart disease.
  • IL-6 we include any natural or synthetic protein with structural and/or functional identity to the human IL-6 protein, such as defined in UniProt Accession No. P05231, or natural variants thereof. IL-6 gene and/or amino acid sequences are disclosed in references 84-86.
  • IL-6 exerts its biological functions via two major pathways: classic signaling and trans-signaling pathways (87). In the classic signaling pathway, IL-6 binds to the IL-6 receptor (IL- 6R) on hepatocytes and some leukocytes. The IL-6 IL-6R complex further recruits the ubiquitously expressed membrane-bound or soluble gpl30 (sgpl30), triggering the dimerization of gpl30 and intracellular signaling.
  • IL-6 receptor IL-6 receptor
  • sgpl30 ubiquitously expressed membrane-bound or soluble gpl30
  • IL-6 interacts with soluble IL-6R (sIL-6R) to form the IL-6 sIL-6R complex, which can bind to gpl30 on any cell and initiate intracellular signaling without a requirement for the stimulated cell to express IL-6R.
  • An antibody which is capable of inhibiting human IL- 6 must be capable of specifically binding to human IL-6, and of inhibiting its interaction with sIL-6R or IL-6R, or otherwise preventing gpl30 activation.
  • capable of specifically binding we include the ability of the antibody or antigen-binding fragment to bind at least 10-fold more strongly to the relevant polypeptide, e.g.
  • IL-6 than to any other polypeptide; preferably at least 50-fold more strongly and more preferably at least 100-fold more strongly.
  • Inhibitory antibodies to IL-6 can typically be divided into two groups; and the putative epitopes on the IL-6 molecule designated Site I and Site II.
  • Site I binders prevent binding to the IL-6R or sIL-6R and thereby prevent gpl30 activation.
  • the Site I epitope was further characterized as comprising regions of both amino terminal and carboxy terminal portions of the IL-6 molecule.
  • Site II- binders prevent gpl30 activation and therefore may recognize a conformational epitope involved in signalling.
  • Binding of the antibody may be measured by surface plasmon resonance, for example, by immobilizing the antibody on a chip and using recombinant human IL-6 as analyte, as described in WO 2004/039826A1.
  • Suitable antibodies may bind IL-6 with an affinity (Kd) of at least IO -9 M, preferably at least 10’
  • binders may be performed by binding to human IL-6-mutant proteins (88). Inhibition of IL-6 activity may be measured by assaying proliferation of the murine B myeloma cell line, 7TD1, in response to IL-6, as described in WO 2004/039826A1. Suitable antibodies may inhibit >50%, such as >90%, such as substantially 100% of 7TD1 cell proliferation in response to IL-6.
  • antibody we include substantially intact antibody molecules, as well as chimeric antibodies, humanised antibodies, human antibodies (wherein at least one amino acid is mutated relative to the naturally occurring human antibodies), single chain antibodies, bi-specific antibodies, antibody heavy chains, antibody light chains, homodimers and heterodimers of antibody heavy and/or light chains, and antigen binding fragments and derivatives of the same.
  • the term also includes antibody-like molecules which may be produced using phage-display techniques or other random selection techniques for molecules.
  • the term also includes all classes of antibodies, including IgG, IgA, IgM, IgD, and IgE.
  • antibody fragments such as Fab, F(ab')2, Fv, Fab', scFv (single-chain variable fragment), or di- scFv and other fragments thereof that retain the antigen-binding site.
  • antibody includes genetically engineered derivatives of antibodies such as single-chain Fv molecules (scFv) and single-domain antibodies (dAbs).
  • Preferred antibodies are chimeric, such as mouse-human chimeric antibodies, CDR- grafted antibodies, humanised antibodies, or human antibodies.
  • the antibody may be a polyclonal antibody, it is preferred if it is a monoclonal antibody, or that the antigen-binding fragment is derived from a monoclonal antibody.
  • Suitable monoclonal antibodies may be prepared by known techniques, for example those disclosed in “Monoclonal Antibodies; A manual of techniques” (89) and in “Monoclonal Hybridoma Antibodies: Techniques and Application” (90).
  • the antibodies may be human antibodies in the sense that they have the amino acid sequence of human antibodies with specificity for the IL-6; however, it will be appreciated that they may be prepared using methods known in the art that do not require immunisation of humans. Suitable antibodies may be prepared from transgenic mice which contain human immunoglobulin loci, as described in "Complete humanization of the mouse immunoglobulin loci enables efficient therapeutic antibody discovery" (91).
  • non-human antibodies can be "humanised” in known ways, for example, by inserting the CDR regions of mouse antibodies into the framework of human antibodies. Chimeric antibodies are discussed in Neuberger et al (92).
  • binding specificity of an antibody or antigen-binding fragment thereof is conferred by the presence of complementarity determining regions (CDRs) within the variable regions of the constituent heavy and light chains.
  • CDRs complementarity determining regions
  • binding specificity for IL-6 is conferred by the presence of one or more and typically all six of the CDR amino acid sequences defined herein.
  • the antibody or antigen-binding fragment comprises an antibody Fc region.
  • the Fc portion may be from an IgG antibody, or from a different class of antibody (such as IgM, IgA, IgD, or IgE).
  • the Fc region may be from an IgGl, IgG2, IgG3, or IgG4 antibody.
  • the Fc region is from an IgGl antibody.
  • the antibody or antigen-binding fragment is an IgG molecule, or is an antigen-binding fragment or variant of an IgG molecule. Suitable antibodies and fragments are described in WO 2004/039826A1.
  • the antibody or fragment which is capable of inhibiting human IL-6 is a chimeric, humanized or CDR grafted antibody or fragment thereof.
  • the antibody or fragment which is capable of inhibiting human IL-6 is a chimeric, humanized or CDR grafted antibody or fragment thereof comprising a heavy chain variable region in which CDR1, CDR2, and CDR3 comprise the amino acid sequences SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO: 3, respectively; and a light-chain variable region in which CDR1, CDR2, and CDR3 comprise the amino acid sequences SEQ ID NO: 4, SEQ ID NO:, 5 and SEQ ID NO: 6, respectively, and a constant region derived from a human IgG antibody.
  • VH CDR2 Glu He Ser Ser Gly Gly Ser Tyr Thr Tyr Tyr Pro Asp Thr Vai Thr Gly (SEQ ID NO. 2)
  • VH CDR3 Gly Leu Trp Gly Tyr Tyr Ala Leu Asp Tyr (SEQ ID NO. 3) VL CDR1 Ser Ala Ser Ser Ser Vai Ser Tyr Met Tyr (SEQ ID NO. 4) VL CDR2 Asp Thr Ser Asn Leu Ala Ser (SEQ ID NO. 5) VL CDR3 Gin Gin Trp Ser Gly Tyr Pro Tyr Thr (SEQ ID NO. 6)
  • the antibody is siltuximab, or an antigen-binding fragment thereof.
  • Siltuximab also known as CNTO328 and CLLB8, with the US FDA UNII Identifier T4H8FMA7IM and the WHO ATC code L04AC11 is a chimeric (human-murine) IgGlK monoclonal antibody that binds to human IL-6.
  • the intact molecule contains 1324 amino acid residues and is composed of two identical heavy chains (approximately 50 kDa each) and two identical light chains (approximately 24 kDa each) linked by inter-chain disulfide bonds.
  • Siltuximab contains the antigen-binding variable region of the murine antibody, CLB-IL-6-8, and the constant region of a human IgGlK immunoglobulin.
  • Siltuximab and methods of preparing it, including by recombinant expression of encoding nucleic acid sequences, are described in WO 2004/039826A1.
  • olokizumab which is a IgG4K antibody humanized from rat (93); elsilimomab (also known as B-E8), which is a mouse IgGlK monoclonal antibody (94); or the human monoclonal antibody clone 1339, which is a high-affinity fully humanized anti-IL-6 monoclonal antibody (IgGl) derived from elsilimomab (95).
  • clazakizumab (formerly ALD518 and BMS-945429), which is an aglycosylated, humanized rabbit IgGl monoclonal antibody against interleukin-6 (96); sirukumab, which is a human monoclonal IgGl kappa antibody (97).
  • Further suitable antibodies include: the MH166 antibody (98); the SK2 antibody (99); Levilimab, which is an anti IL-6 monoclonal antibody initially developed to treat rheumatoid arthritis (100); and ARGX-109, which is a preclinical stage human antibody candidate developed by arGEN-X from its SIMPLE AntibodyTM platform and which is said to have outstanding neutralization potency for IL-6. Fragments of any of these antibodies may also be used.
  • the antibody or fragment should be prepared under sterile conditions.
  • the appropriate volume of antibody or fragment should be withdrawn from the vials. It is recommended that the antibody solution is filtered (0.2 to 1.2 pm) before injection into the patient either by using an in-line filter during infusion or by filtering the solution with a particle filter (e.g., filter Nr. MF1830, Impromediform, Germany).
  • the volume of the antibody is typically added to an infusion bag containing 5% dextrose.
  • Siltuximab is available as a single-use vial containing 100 mg or 400 mg siltuximab powder for concentrate for solution for infusion, and should be stored at refrigeration temperature.
  • the siltuximab powder is typically provided with one or more excipients, typically histidine, histidine hydrochloride monohydrate, polysorbate 80, and sucrose. After reconstitution with single-use sterile water for injection, the solution contains 20 mg siltuximab per mL.
  • excipients typically histidine, histidine hydrochloride monohydrate, polysorbate 80, and sucrose. After reconstitution with single-use sterile water for injection, the solution contains 20 mg siltuximab per mL.
  • Antibodies or fragments may be formulated in other ways, as known in the art.
  • the dose of the antibody when administered by intravenous administration, such as by infusion, may be 11 ⁇ 3 mg/kg patient body weight, optionally 11 mg/kg.
  • a suitable dose of the fragment is a dose having an equivalent antagonistic effect on human IL- 6.
  • the combination therapy of the present invention partly comprises administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6, not human IL-6R.
  • inhibitors of human IL6 will be advantageous over inhibitors of IL-6R for treating AMR in lung transplant recipients for many reasons.
  • treatment with monoclonal antibodies to IL-6R may result in significant increases in IL-6 and IL-6R levels (44, 65). Although this may not cause deleterious effects while treatment is ongoing, there is a theoretical possibility that a rebound IL- 6 effect may be seen when treatment is discontinued. Indeed, the occurrence of allograft loss in 4 kidney recipients with chronic AMR who stopped Tocilizumab supports this possibility (50, 56). Furthermore, the inventors consider that considerably lower doses of monoclonal antibodies to IL-6 may be used compared to those to IL-6R because soluble IL-6R is present at high concentrations compared to IL-6, and all soluble IL-6R have to be saturated with a therapeutic antibody to exert its pharmacodynamic effect (66).
  • IL-6R binds not only IL-6 but also IL-27p28, so the blockade of IL-6R may affect other signaling pathways (67).
  • Siltuximab IL6 inhibitor
  • IL-6R inhibitor IL-6R inhibitor
  • IL-6R inhibitor IL-6R inhibitor
  • the antibody or fragment which is capable of inhibiting human IL-6 is selected from siltuximab, olokizumab, elsilimomab, mAb 1339, clazakizumab, sirukumab, levilimab and ARGX- 109.
  • the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab.
  • Siltuximab has been used in patients with hematologic diseases/malignancies, solid tumors and MCD.
  • Phase 1 studies that included patients with advanced solid tumors, refractory multiple myeloma, or MCD, common grade > 3 adverse events (AE) included leukopenia, neutropenia, thrombocytopenia, hypercholesterolemia, anemia, fatigue, infections, and abnormal liver enzymes (62, 69-71).
  • AE adverse events included leukopenia, neutropenia, thrombocytopenia, hypercholesterolemia, anemia, fatigue, infections, and abnormal liver enzymes (62, 69-71).
  • Phase 2 studies in MCD Siltuximab has generally been well-tolerated during long-term follow-up, with a similar pattern of grade > 3 AEs as in Phase 1 studies (72- 74).
  • Siltuximab's safety data are consistent with those of other IL-6/IL-6R monoclonal antibodies (64).
  • Serious infections are the most common serious adverse events (SAE) in clinical trials and post-marketing surveillance studies; other SAE include GI tract perforation, hepatitis, and pancreatitis (64).
  • Siltuximab dosing studies for indications other than AMR have examined different doses ranging between 3 mg/kg to 15 mg/kg with most doses given every 2 or 3 weeks in patients with hematologic diseases/malignancies, solid tumors, or Multicentric Castleman's Disease (62, 70, 71, 75,76). Measuring IL-6 concentrations accurately during treatment with Siltuximab is not feasible because Siltuximab-neutralized IL-6 complexes distort quantification methods (76). Thus, levels of C-reactive protein (CRP) are used as a surrogate for IL-6 activity because IL-6 is the primary factor that drives CRP production by hepatocytes (76, 77).
  • CRP C-reactive protein
  • Immunosuppressive therapy we include the standard, routine, common immunosuppressive therapies which are given to transplant recipients before, after or during the transplant procedure to suppress their immune system and reduce the risk of the recipient's body rejecting the donor lung(s) (transplant). The purpose is to stop an unwanted immune response that would damage survival of the transplanted tissue and/or of the subject. Some transplant recipients may be on a life-long immunosuppressive treatment regimen after the transplant. Some patients may be able to reduce or stop taking certain immunosuppressive therapies (e.g., corticosteroids) after the lung transplant, which may reduce the risk of the side effects and complications associated with the immunosuppressive therapies. This is particularly because continuation of immunosuppressive therapies may result in immunodeficiency, by which the subject may be susceptible to opportunistic infection and/or increase the likelihood of cancer due to reduced surveillance of damaged cells by the immune system.
  • immunosuppressive therapies e.g., corticosteroids
  • Some common immunosuppressive therapy options administered to patients after a lung transplant include glucocorticoids (corticosteroids), monoclonal antibodies (mAbs), nucleotide blocking agents, calcineurin inhibitors, and mTOR inhibitors.
  • glucocorticoids corticosteroids
  • monoclonal antibodies monoclonal antibodies
  • nucleotide blocking agents calcineurin inhibitors
  • calcineurin inhibitors calcineurin inhibitors
  • mTOR inhibitors a common immunosuppressive therapy options administered to patients after a lung transplant.
  • monoclonal antibodies are known immunosuppressants
  • antibodies or fragments which are capable of inhibiting human IL-6 are not known for use in treatment of AMR in lung transplant patients.
  • Suitable immunosuppressive therapy options are known to the person skilled in the art.
  • the immunosuppressive therapy may comprise a multi-drug regimen.
  • the combination therapy may comprise two or more of the immunosuppressive therapy treatments described herein.
  • the immunosuppressive therapy may comprise administering one or more of a baseline immunosuppression maintenance therapy, and/or a standard of care treatment for AMR.
  • Immunosuppressive therapies may be categorised as induction agents, maintenance therapy, or rescue therapy (i.e. to treat rejection when observed).
  • Induction agents are administered at the time of transplant and are generally more powerful immunosuppressive agents; for this reason they are given for a short acute period.
  • Maintenance therapy comprises medications used over a longer period.
  • Rescue therapy is given when clinical signs of rejection require additional action, such as when the transplant rejection becomes refractory to other medications.
  • “baseline immunosuppression maintenance therapy” is defined as immunosuppressive therapy that is categorised as maintenance therapy and used as a form of therapy to be administered chronically to ensure and improve tolerance and continued acceptance of the recipient organ or graft.
  • calcineurin inhibitors cyclosporine and tacrolimus
  • mammalian target of rapamycin (mTOR) inhibitors sirolimus and everolimus
  • antiproliferative agents azathioprine and mycophenolic acid
  • costimulatory blockers e.g., cowatacept, cowatacept, cowatacept, cowatacept, cowatacept, cowatacept, cowatacept, and corticosteroids.
  • lung transplant recipients may be treated with a combination of Tacrolimus, Mycophenolate Mofetil (MMF), and Prednisone (or methylprednisolone) for maintenance immunosuppression.
  • MMF Mycophenolate Mofetil
  • Prednisone or methylprednisolone
  • Cyclosporine A is substituted for Tacrolimus or Azathioprine is substituted for MMF because of toxicity.
  • patients may be administered a routine maintenance immunosuppression therapy as described herein.
  • the baseline immunosuppression maintenance therapy may comprise one or more of tacrolimus, cyclosporine A, Mycophenolate Mofetil (MMF), azathioprine, and prednisone.
  • the baseline immunosuppression maintenance therapy may comprise:
  • the standard of care treatment for AMR is defined as a routine or standard treatment for pulmonary AMR that is commonly or generally administered to a patient before, during or after receiving a lung transplant.
  • this may include preventative immunosuppressive treatment administered before, during or after transplantation of one or more lung(s) or lobe(s) thereof, to prevent AMR developing, and/or routine immunosuppressive treatment administered before, during or after transplantation to ameliorate, stabilise, improve, or (partially) treat AMR in a lung transplant recipient.
  • the standard of care treatment for AMR may comprise one or more of a corticosteroid, intravenous immune globulin (IVIG), Rituximab, Bortezomib, Carfilzomib, antithymocyte globulin (ATG), plasma exchange (PLEX), tacrolimus, cyclosporine A, Mycophenolate Mofetil (MMF), azathioprine, prednisone, and methylprednisolone, and optionally acetaminophen (paracetamol) and/or diphenhydramine.
  • IVIG intravenous immune globulin
  • Rituximab Rituximab
  • Bortezomib Bortezomib
  • Carfilzomib antithymocyte globulin
  • PLEX antithymocyte globulin
  • tacrolimus cyclosporine A
  • MMF Mycophenolate Mofetil
  • azathioprine prednisone
  • the corticosteroid may be selected from one or more of prednisolone, prednisone, methylprednisolone, cortisone, dexamethasone, betamethasone and hydrocortisone. These are all commercially available corticosteroids.
  • the immunosuppressive therapy may comprise acetaminophen, diphenhydramine, and methylprednisolone.
  • the acetaminophen, diphenhydramine, and methylprednisolone may be administered 30- 60 minutes prior to an or each administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6.
  • the antibody or fragment which is capable of inhibiting human IL-6 may be siltuximab.
  • the administration of the immunosuppressive therapy may comprise the administration of acetaminophen 650 or 1000 mg by mouth (oral administration e.g, by tablet or capsule), diphenhydramine 25 or 50 mg by mouth (oral administration e.g, by tablet or capsule), and methylprednisolone 60 mg intravenously.
  • antibody or fragment which is capable of inhibiting human IL-6 is preferably Siltuximab.
  • the acetaminophen, diphenhydramine, and methylprednisolone may be administered 30-60 minutes prior to an or each administration of the antibody of fragment (e.g., Siltuximab).
  • acetaminophen, diphenhydramine, and methylprednisolone do not necessarily need to be administered with every dose of the antibody or fragment but they may be administered with each dose of antibody or fragment (e.g., Siltuximab) if required.
  • the skilled person would be able to design a suitable treatment plan of the immunosuppressive therapy according to the needs of the patient.
  • the treatment plan may be revised during the treatment (e.g. between doses of the antibody or fragment) as the patient requires (for example in reaction to the patient's toleration of a particular dose or type of antibody or fragment, or to take into account the perceived efficacy of treating AMR in a particular patient a particular dose or type of antibody or fragment).
  • the antibody or fragment which is capable of inhibiting human IL-6 may be administered via the same or a different administration route as the immunosuppressive therapy.
  • the antibody or fragment which is capable of inhibiting human IL-6 may be prepared e.g. for parenteral administration e.g., subcutaneous, intramuscular, intravenous, intra-dermal, intra-articular or periarticular administration, particularly in the form of liquid solutions or suspensions; or for inhalation to the lungs e.g. pulmonary administration, particularly in the form of solutions, suspensions including nanosuspensions for nebulisation, or suspension or solution pressurised or non-pressurised aerosols.
  • the immunosuppressive therapy may be prepared for parenteral administration e.g., subcutaneous, intramuscular, intravenous, intra-dermal, intra-articular or peri-articular administration, particularly in the form of liquid solutions or suspensions; for oral administration, particularly in the form of tablets, capsules, powder, granules, solid dispersions or in the form of liquid solutions or suspensions including nanosuspensions; for inhalation to the lungs or nose e.
  • pulmonary or intranasal administration particularly in the form of dry powders, solutions, suspensions including nanosuspensions for nebulisation, nasal sprays or drops comprising solutions or suspensions or suspension or solution pressurised or non-pressurised aerosols; for topical or transdermal administration e.g. as creams, sprays, foams, gels, ointments, liquids, patches; for mucosal administration e.g. to buccal, sublingual or vaginal mucosa, and for rectal administration e.g. in the form of a foam or suppository.
  • the antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy may be administered by inhalation.
  • inhaled medications are their direct delivery to the area of rich blood supply in comparison to many medications taken by oral route. Thus, the absorption is very rapid as the alveoli have an enormous surface area and rich blood supply and first pass metabolism is bypassed.
  • the antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy may conveniently be administered in unit dosage form and may be prepared by any of the methods well-known in the pharmaceutical art.
  • the antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy may also conveniently be administered in multiple unit dosage form.
  • the present invention also provides an inhalation device containing the antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy of the present invention.
  • said device is a metered dose inhaler (MDI), which contains a pharmaceutically acceptable chemical propellant to push the medication out of the inhaler.
  • MDI metered dose inhaler
  • the immunosuppressive therapy may be administered by intranasal administration.
  • the nasal cavity's highly permeable tissue is very receptive to medication and absorbs it quickly and efficiently.
  • Nasal drug delivery is less painful and invasive than injections, generating less anxiety among patients. By this method absorption is very rapid and first pass metabolism is usually bypassed, thus reducing inter-patient variability.
  • the immunosuppressive therapy may be administered by transdermal administration.
  • transdermal and transmucosal patches, creams, ointments, jellies, solutions or suspensions may be employed.
  • the immunosuppressive therapy may be administered by sublingual administration.
  • the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is by intravenous administration, optionally by infusion, optionally wherein the infusion is over the course of one hour.
  • compositions comprising IL-6 inhibitors and other pharmaceutical compositions for administering immunosuppressive therapy.
  • Such compositions may comprise a prophylactically or therapeutically effective amount of the active drug (the IL-6 inhibitor and/or a drug required by the immunosuppressive therapy), and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a particular carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • compositions in the context of the therapies and administrations described herein can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical agents are described in "Remington's Pharmaceutical Sciences.”
  • Such compositions will contain a prophylactically or therapeutically effective amount of the agent, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration, which can be oral, intravenous, intraarterial, intrabuccal, intranasal, nebulized, bronchial inhalation, intra-rectal, vaginal, topical or delivered by mechanical ventilation.
  • Pharmaceutically acceptable salts include the acid salts and those which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine,
  • compositions of the disclosure are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water-free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the immunosuppressive therapy is intended to be a routine (standard) immunosuppressive therapy generally administered to lung transplant recipients
  • a person skilled in the art would be aware of the appropriate administration routes for different immunosuppressive therapies.
  • the administration route may vary inter alia according to patient characteristics, dosage amount(s) and/or frequency, and/or the type of immunosuppressive therapy.
  • the administration route may be different across different immunosuppressive therapies within the multi-drug regimen.
  • the antibody or fragment which is capable of inhibiting human IL-6 may be administered according to a dosage regimen.
  • the administration may include one or more (including two or more, three or more, four or more) doses of the antibody or fragment.
  • the administration may comprise one, two, three, four, five, six, seven, eight, nine or more doses to a subject of the antibody or fragment which is capable of inhibiting human IL-6.
  • the antibody or fragment which is capable of inhibiting human IL-6 is administered as 2 or more doses.
  • the antibody or fragment which is capable of inhibiting human IL-6 may be administered as 2 or 3 doses.
  • the antibody or fragment which is capable of inhibiting human IL-6 may be administered as 2 doses.
  • the antibody or fragment which is capable of inhibiting human IL-6 may be administered as 3 doses.
  • the dose of the antibody or fragment is determined according to the weight in kg of the patient. Where a dose is given in the units of "mg/kg", this refers to mg of antibody or fragment or other drug per kg of body weight of the patient.
  • An antibody fragment is to be administered at an equivalent fragment dose having an equivalent antagonistic effect on human IL-6 to the whole antibody from which the fragment is derived.
  • the equivalent fragment dose may be calculated according to the fragment molecular weight compared to the molecular weight of the whole antibody, also referred to as parent antibody.
  • a fragment dose that is one third of the antibody dose should provide an equivalent antagonistic effect on human IL-6.
  • the equivalent fragment dose for the Fab fragment would be 4 mg/kg.
  • IL-6 may also be determined according to the amount of human IL-6 that the fragment can specifically bind to, compared to the amount of human IL-6 that the parent antibody can specifically bind to. These amounts may be determined by various assays, including ELISA.
  • the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is in a dose of 1 mg/kg to 15 mg/kg, such as 1 mg/kg, 1.5 mg/kg, 2 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, 5 mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg,
  • the dose may be 5.5 mg/kg or 11 mg/kg.
  • the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab. More preferably, the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab, and is administered in a dose of 5.5 mg/kg or 11 mg/kg.
  • Particular dosage regimens of the antibody or fragment which is capable of inhibiting human IL-6 include:
  • a first dose of the antibody or fragment preferably siltuximab
  • a one dose treatment i.e., a one dose treatment
  • each dose may be from 1 mg/kg to 15 mg/kg.
  • each dose may be 5.5 mg/kg or 11 mg/kg.
  • Each dose administered in the same dosage regimen does not have to be of the same dosage amount.
  • a subject may be administered a first dose of 11 mg/kg of the antibody or fragment (preferably siltuximab) on day 1 of treatment, and then administered a second dose of 5.5 mg/kg of the antibody or fragment (preferably siltuximab) on day 8 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 11 mg/kg on day 8 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 11 mg/kg on day 22 ⁇ 3 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 11 mg/kg on day 22 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment
  • a second dose of siltuximab is administered at 11 mg/kg on day 8 of treatment
  • a third dose of siltuximab is administered at 11 mg/kg on day 22 ⁇ 3 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment
  • a second dose of siltuximab is administered at 11 mg/kg on day 8 of treatment
  • a third dose of siltuximab is administered at 11 mg/kg on day 22 of treatment.
  • a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment.
  • a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment
  • a second dose of siltuximab is administered at 5.5 mg/kg on day 22 ⁇ 3 of treatment.
  • a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment.
  • a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment
  • a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment
  • a third dose of siltuximab is administered at 5.5 mg/kg on day 22 ⁇ 3 of treatment.
  • a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 22 ⁇ 3 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22 ⁇ 3 of treatment.
  • a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment.
  • the subject may be administered a different antibody or fragment which is capable of inhibiting human IL-6 as part of the same dosing regime.
  • the first dose may comprise siltuximab
  • the second dose may comprise a different antibody or fragment which is capable of inhibiting human IL-6, such as one or more IL-6 inhibitors selected from olokizumab, elsilimomab, mAb 1339, clazakizumab, sirukumab, levilimab, SK2, MH166, and ARGX-109.
  • a third dose may comprise the same antibody or fragment as the first and/or second dose, or a different antibody or fragment as the first and/or second dose.
  • each dose comprises or consists of siltuximab.
  • the treatment of the present invention is a combination therapy of (i) an antibody or fragment which is capable of inhibiting human IL-6; and (ii) an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
  • the immunosuppressive therapy does not comprise any of siltuximab, olokizumab, elsilimomab, mAb 1339, clazakizumab, sirukumab, levilimab, SK2, MH166, and ARGX- 109.
  • the immunosuppressive therapy may comprise IL6R inhibitors such as Tocilizumab.
  • the only antibody inhibitor of IL-6 signalling will be the antibody or fragment which is capable of inhibiting human IL-6.
  • the immunosuppressive therapy would not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6 or human IL-6R.
  • the only inhibitor of IL-6 signaling will be the antibody or fragment which is capable of inhibiting human IL-6.
  • the immunosuppressive therapy would not comprise any drug which is capable of inhibiting human IL-6 or human IL-6R.
  • the combination therapy may comprise (i) an antibody or fragment which is capable of inhibiting human IL-6; and (ii) an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise the antibody or fragment which is capable of inhibiting human IL-6 from (i).
  • the immunosuppressive therapy of (ii) does comprise not Siltuximab.
  • each or a dose of (i) an antibody or fragment which is capable of inhibiting human IL-6 may be administered with or without a dose of (ii) immunosuppressive therapy, as long as a dose of (ii) immunosuppressive therapy is administered at least once during the treatment.
  • the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is given in a first treatment dose initiated within the period 24 hours before or after initiation of the immunosuppressive therapy.
  • the administration of the first treatment dose of the antibody of fragment may be initiated within the period 2 hours before or after initiation of the immunosuppressive therapy.
  • the administration of the first treatment dose of the antibody of fragment may be initiated within the period one hour before or after initiation of the immunosuppressive therapy.
  • the administration of the first treatment dose of the antibody of fragment may be initiated substantially at the same time as initiation of the immunosuppressive therapy.
  • the immunosuppressive therapy is administered 30 to 60 minutes prior to an or each administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6.
  • the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab.
  • the immunosuppressive therapy is administered 30 to 60 minutes prior to one or more doses (particularly the first dose, or the only dose if there is only one dose administered) of the antibody or fragment (preferably Siltuximab).
  • the immunosuppressive therapy for example may be administered 30 to 60 minutes prior to every dose, only the first dose, only the last dose, only the second dose, only one dose, only one or two doses out of a three dose treatment, every other dose, every second dose, or every third dose and so on, of the antibody or fragment (for example, Siltuximab).
  • the administration of the combination therapy may further comprise administration to the subject of a prophylactic medication, wherein the prophylactic medication is an antiviral and/or antimicrobial medication.
  • the prophylactic medication may be one or more of an antiviral medication, or an antimicrobial medication.
  • any suitable antiviral or antimicrobial medication may be administered prophylactically, i.e. to prevent possible viral or microbial infection.
  • microbial infection we include bacterial, mycobacterial and fungal infections, such that an antimicrobial medication may be an antibiotic agent against bacteria or mycobacteria, or an antifungal agent against fungi (particularly yeasts or molds).
  • the prophylactic medication may comprise one or more of Valganciclovir, Trimethoprim-Sulfamethoxazole, Voriconazole, Posaconazole, and Isavuconazonium.
  • prophylactic medications are considered standard treatments for lung transplant recipients (i.e., these medications are part of the standard of care), and the skilled person would be able to determine the appropriate dosage, medication, and prophylactic treatment regimen. These prophylactic medications are known and commercially available.
  • Participants at risk for cytomegalovirus may be treated with Valganciclovir prophylactically after treatment for AMR.
  • all lung transplant recipients may be maintained on prophylaxis for Pneumocystis jirovecii with either Trimethoprim-Sulfamethoxazole or an alternative drug if allergic to sulfa.
  • All patients being evaluated for AMR may undergo bronchoscopy with lung biopsies, bronchial washings, and/or bronchoalveolar lavage as part of the routine clinical evaluation.
  • Bronchoscopy specimens may be routinely tested for mycobacterial, fungal, and bacterial organisms as well as polymerase chain reaction (PCR) testing for CARV.
  • PCR polymerase chain reaction
  • positive mycobacterial, fungal, or bacterial cultures do not necessarily represent invasive disease or infection, patients may be routinely started on appropriate antimicrobial therapy before initiating treatment for AMR to minimize the risk of invasive disease and infection after treatment for AMR.
  • Patients who have positive cultures for a mold from bronchoscopy specimens may be routinely treated with Voriconazole, Posaconazole, or Isavuconazonium. Patients who have a positive bacterial culture may be treated with appropriate antimicrobials. For patients who have a positive mycobacterial culture, anti-mycobacterial therapy may be initiated depending on the specific organism.
  • Figure 1 shows (A) serum IL-6 concentrations in a cohort of lung transplant patients with AMR compared to healthy non-transplant individuals, and (B) immunofluorescence staining for IL-6 in lung biopsies of patients with AMR.
  • Figure 2 shows the traditional 3+3 Phase 1 clinical trial design used in Example 2.
  • Figure 3 shows the dosing and follow-up timelines for Cohorts A-D in Example 2.
  • Example 1 Measurements of IL-6 in patients with AMR
  • the secondary objectives are:
  • the primary endpoint is safety and tolerability which will be assessed based on the development of dose limiting toxicities (DLT), adverse events (AE), and serious adverse events (SAE).
  • DLT dose limiting toxicities
  • AE adverse events
  • SAE serious adverse events
  • Allograft survival defined as death or undergoing re-transplantation.
  • DSA Donor-specific antibodies to human leukocyte antigens
  • DSMB Data and Safety Monitoring Board
  • Allergic reaction An allergic reaction is defined as a disorder characterized by an adverse local or general response from exposure to an allergen. Grade 3 allergic reaction is one that results in bronchospasm, requires hospitalization, or intravenous intervention.
  • Antibody-mediated rejection will be defined based on the
  • Bronchoalveolar lavage is medical procedure performed during bronchoscopy in which a small scope is inserted into a patient's nose or mouth and passed into the lungs. Once in the lungs, sterile saline is instilled, coating the lining the lungs, and then quickly aspirated out of the lungs.
  • DSA Clearance of donor-specific antibodies
  • DSA Clearance of donor-specific antibodies
  • DLT Death is defined as a Dose Limiting Toxicity if the CEAC adjudicates that death was due to infection.
  • Donor-specific antibodies are antibodies to donor human leukocyte antigens (HLA) with a Mean Fluorescence Intensity > 1000. These can be either pre-existing or de novo.
  • HLA human leukocyte antigens
  • Hepatitis as a DLT: Hepatitis is defined as a Dose Limiting Toxicity if the AST or ALT > 5 times the upper limit of normal or if the total bilirubin > 3 times the upper limit of normal.
  • Hyperlipidemia as a DLT: Hyperlipidemia is defined as a Dose Limiting Toxicity if it results in pancreatitis, requires hospitalization, or prolongs a hospitalization.
  • Hypersensitivity reaction A hypersensitivity reaction is defined as a Dose Limiting Toxicity if this meets the definition of a Grade > 3 allergic reaction.
  • Infection as a DLT Infections including CMV, fungemia, and respiratory tract infections are defined as Dose Limiting Toxicities if they require hospitalization or prolong a hospitalization.
  • Neutropenia as a DLT: Neutropenia is defined as a Dose Limiting Toxicity if the absolute neutrophil count ⁇ 500 cells/mm 3 for at least 3 days.
  • Opportunistic infections are infections that occur more often or are more severe in immunocompromised patients. These include tissue invasive cytomegalovirus, varicella zoster, tuberculosis, Pneumocystis jirovecii, nocardia, endemic mycoses, and toxoplasmosis.
  • Probable CLAD is defined as a >20% decline in FEVi compared to the baseline value on 2 measurements at least 3 weeks apart and after exclusion or adequate treatment of potential secondary causes of allograft dysfunction (e.g., respiratory infection, acute cellular rejection).
  • Pruritis as a DLT: Pruritis is defined as a Dose Limiting Toxicity if it is widespread and constant, limits self-care, or requires systemic steroids.
  • Rash as a DLT: Rash is defined as a Dose Limiting Toxicity if it covers > 30% of the body surface area with moderate or severe symptoms limiting self-care.
  • Thrombocytopenia is defined as a Dose Limiting Toxicity if the platelet count ⁇ 50,000 cells/mm3 for at least 3 days.
  • Cohort A 3 participants will be treated with Siltuximab 5.5 mg/kg for 2 doses on days 1 and 8 (Figure 3). If none of the 3 participants in Cohort A experiences a DLT (Table 3), the next 3 participants will be enrolled in Cohort B (see below). However, if 1 participant in Cohort A experiences a DLT, 3 more participants will be added to Cohort A and treated with the same dose ( Figure 2). If no additional participants experience a DLT in Cohort A, the study will progress to Cohort B. If 2 or more of the 6 participants or 2 of the initial 3 participants in Cohort A experience a DLT, the next 3 participants will be enrolled in Cohort A2.
  • Cohort A2 3 participants will be treated with 5.5 mg/kg for 2 doses on days 1 and 22. If none of the 3 participants in Cohort A2 experiences a DLT, the study will progress to Cohort B. If 1 participant in Cohort A2 experiences a DLT, 3 more participants will be added to Cohort A2 and treated with the same dose.
  • Cohort B 3 participants will be treated with Siltuximab 11 mg/kg for 2 doses on days 1 and 22 (Figure 3). If none of the 3 participants in Cohort B experiences a DLT, the next 3 participants will be enrolled in Cohort C. If 1 participant in Cohort B experiences a DLT, 3 more participants will be added to Cohort B and treated with the same dose. If no additional participants experience a DLT in Cohort B, the study will progress to Cohort C. If 2 or more of the 6 participants or 2 of the initial 3 participants in Cohort B experience a DLT, the maximum tolerated dose (MTD) will be designated as that used in Cohort A (or Cohort A2 if that was the previous Cohort before Cohort B).
  • MTD maximum tolerated dose
  • Cohort C In Cohort C, 3 subjects will be treated with Siltuximab 11 mg/kg for 2 doses on days 1 and 8 ( Figure 3). If none of the 3 participants in Cohort C experiences a DLT, the next 3 participants will be enrolled in Cohort D. If 1 participant in Cohort C experiences a DLT, 3 more participants will be added to Cohort C and treated with the same dose. If no additional participants experience a DLT in Cohort C, the study will progress to Cohort D. If 2 or more of the 6 participants or 2 of the initial 3 participants in Cohort C experience a DLT, the MTD will be designated as that used in Cohort B.
  • Cohort D 3 subjects will be treated with Siltuximab 11 mg/kg on days 1, 8, and 22 for a total of 3 doses (Figure 3). If none of the 3 participants in Cohort D experiences a DLT, we will designate this as the MTD. If 1 participant in Cohort D experiences a DLT, 3 more participants will be added to Cohort D and treated with the same dose. If no additional participants experience a DLT, we will consider this the MTD. If 2 or more of the 6 participants or 2 of the 3 initial participants in Cohort D experience a DLT, the MTD will be designated as that used in Cohort C.
  • the MTD The MTD
  • the MTD is the highest dosing schedule where ⁇ 33% of patients experience a DLT. Once the MTD is identified, we will enrol additional patients to reach a total number of 18 (in all cohorts) and treat them with this dosing schedule to confirm MTD and develop preliminary data to inform the design of a future Phase 2 clinical trial.
  • the approved dose of Siltuximab for MCD is 11 mg/kg intravenously every 3 weeks.
  • the optimal dose and dosing schedule for the treatment of AMR after LT are not known.
  • the role of IL-6 and its concentrations in serum and local tissues is different in different disease states. Indeed, IL-6 concentrations in serum are generally higher in MCD than in AMR.
  • the primary endpoint is safety and tolerability which will be assessed based on the development of dose limiting toxicities (DLT), adverse events (AE), and serious adverse events (SAE). Table 3 below details and defines the different DLT.
  • DLT dose limiting toxicities
  • AE adverse events
  • SAE serious adverse events
  • DLT The development of DLT will be assessed during the DLT follow-up period which is defined as 30 days after the last dose of Siltuximab (Figure 3). All DLT, AE, and SAE will be reviewed by the CEAC to adjudicate if a DLT has occurred. Where applicable, the study will use the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0.
  • CTCAE Common Terminology Criteria for Adverse Events
  • the study defines a hypersensitivity reaction as a DLT if this meets CTCAE Grade > 3 allergic reaction.
  • An allergic reaction is defined as a disorder characterized by an adverse local or general response from exposure to an allergen. Grade 3 allergic reaction is one that results in bronchospasm, requires hospitalization, or intravenous intervention.
  • DLT Death is defined as a DLT if the CEAC adjudicates that death was due to infection because approximately 20% of LT recipients with AMR die within 30 days of the diagnosis without treatment with IL-6 signaling blockade (20).
  • Grade 3 thrombocytopenia platelet count ⁇ 50,000 cells/mm3
  • Grade 4 neutropenia absolute neutrophil count ⁇ 500 cells/mm3 lasting at least 3 days are defined as DLT.
  • Grade 3 rash is defined as a DLT if it covers > 30% of the body surface area (BSA) with moderate or severe symptoms limiting self-care.
  • BSA body surface area
  • Grade 3 pruritis is defined as a DLT if it is widespread and constant, limits self-care, or requires systemic steroids.
  • Hyperlipidemia is defined as a DLT if it results in pancreatitis or requires hospitalization or prolongs a hospitalization. Infections are defined as DLT if they require hospitalization or prolong a hospitalization.
  • requirement for intravenous antibiotics is the threshold for Grade 3 infections in the CTCAE.
  • LT recipients are frequently treated with intravenous antibiotics for non-serious upper or lower respiratory tract infections with antibiotic-resistant organisms. Yet, the use of intravenous antibiotics in this setting is not an indication of the clinical severity of infection. Thus, we have modified the definition of infection as a DLT to be more applicable to LT recipients.
  • the study will enrol adult lung transplant recipients who develop probable or definite AMR according to the 2016 ISHLT definition (23). The study will not enrol children as the lung transplant programs enrolling participants do not provide care for children. Lung transplant recipients who develop AMR have a poor prognosis. IL-6 plays an important role in mediating allograft injury, and IL-6 blockade might mitigate this. The eligibility criteria for enrolment have been determined based on known adverse events associated with IL-6 blockade and Siltuximab, the package insert, and the risk of infection associated with augmented immunosuppression.
  • Siltuximab is the investigational agent in this study. Details describing the study related product, labeling, supply, storage, preparation, dispensing, monitoring and accountability are described below. Siltuximab is manufactured by EUSA Pharma and is approved by the FDA as detailed below. Siltuximab is a first-in-class chimeric (humanmouse) immunoglobulin G1K (IgGlK) monoclonal antibody against human IL-6 in a Chinese hamster ovary cell line.
  • IgGlK immunoglobulin G1K
  • the Siltuximab dose is given as an intravenous infusion over 60 minutes. This Phase 1 study will assess the safety and tolerability of 4 dosing schedules to determine the MTD (see Section 4.1 above and Figure 3). Complete blood count should be checked before each dose of Siltuximab. Before the first administration, the absolute neutrophil count must be > 1,000 cells/mm 3 , the platelet count must be > 75,000 cells/mm 3 , and the hemoglobin must be ⁇ 17 g/dL. Before subsequent doses, the absolute neutrophil count must be > 1,000 cells/mm 3 , the platelet count must be > 50,000 cells/mm 3 , and the hemoglobin must be ⁇ 17 g/dL. If these criteria are not me, the dose may be delayed but the dose should not be reduced. Participants will be premedicated 30-60 minutes prior to the start of the Siltuximab infusion using:
  • Siltuximab does not contain preservatives. Do not store any unused portion of the reconstituted product or of the infusion solution. Waste material should be disposed of in accordance with local requirements.
  • Siltuximab will be administered in a hospital or outpatient infusion center where full resuscitation facilities are immediately available and under close supervision of the investigator or designated safety accessor.
  • the intravenous infusion will be supervised by the clinical staff (e.g., clinical nurse or physician) at the participating institutions.
  • a history of each infusion and any adverse side effects will be recorded and reported using appropriated case report forms (CRF).
  • clinical labs Prior to each infusion, clinical labs (ANC, platelets, bilirubin, ALT, and AST) will be completed. Results must be available prior to initiating the infusion.
  • Vital signs (temperature, blood pressure, pulse, and respiratory rate) will be obtained prior to the start of each infusion and every 15 minutes until the end of the infusion, and every hour thereafter for a total of 2 hours after the first infusion, and 1 hour after subsequent infusions. After the infusion, the intravenous line should remain in the participant for at least 1 hour to enable the administration of drugs if necessary. Additional vital signs may be obtained as clinically indicated. 6.5. Instructions regarding siltuximab dosing delays and discontinuation criteria
  • Siltuximab Closely monitor patients for the development of signs and symptoms of infection during and after treatment with Siltuximab, as signs and symptoms of acute inflammation may be lessened due to suppression of the acute phase reactants. Hold Siltuximab if a patient develops a serious infection, an opportunistic infection, or sepsis. A patient who develops a new infection during treatment with Siltuximab should undergo a prompt and complete diagnostic workup appropriate for an immunocompromised patient, initiate appropriate antimicrobial therapy, and closely monitor the patient. Siltuximab may be resumed at the next scheduled dose timepoint if the infection has resolved.
  • Treatment with Siltuximab was associated with a higher incidence of neutropenia. Infections have been uncommonly reported in association with treatment-related neutropenia in studies and post marketing clinical experience. Per package insert, it is not recommended to initiate Siltuximab in patients with an ANC ⁇ 1000 cells/mm 3 . If the ANC is ⁇ 1000 cells/mm 3 , treatment with Siltuximab may be delayed, but the dose should not be reduced. For this study, if the ANC is ⁇ 1000 cells/mm 3 , the scheduled dose of Siltuximab may be delayed up to 3 days. If during this time the ANC is > 1000 cells/mm 3 , the dose may be given. However, if the ANC remains ⁇ 1000 cells/mm 3 , the dose will not be administered.
  • Treatment with Siltuximab was associated with a reduction in platelet count. Treatment-related reduction in platelets was not associated with serious bleeding events in clinical trials. Per the package insert, it is not recommended to initiate Siltuximab in patients with a platelet count ⁇ 75,000 cells/mm 3 , and it is not recommended to receive subsequent doses of Siltuximab in patients with a platelet count ⁇ 50,000 cells/mm 3 . For this study, if the platelet count is ⁇ 50,000 cells/mm 3 , the scheduled dose of Siltuximab may be delayed up to 3 days. If during this time the platelet count is > 50,000 cells/mm 3 , the dose may be given. However, if the platelet count remains ⁇ 50,000 cells/mm 3 , the dose will not be administered.
  • Siltuximab may increase hemoglobin levels in patients with MCD. Treatment with Siltuximab is not recommended in patients with a hemoglobin > 17 g/dL. For this study, if the hemoglobin is > 17 g/dL, the scheduled dose of Siltuximab may be delayed up to 3 days. If during this time the hemoglobin is ⁇ 17 g/dL, the dose may be given. However, if the hemoglobin remains > 17 g/dL, the dose will not be administered.
  • the site PI believes that study therapy is no longer in the best interest of the participant (e.g., due to medical decision, pneumonia, serious infection, opportunistic infection).
  • lung transplant recipients are treated with a combination of Tacrolimus, Mycophenolate Mofetil (MMF), and Prednisone for maintenance immunosuppression.
  • MMF Mycophenolate Mofetil
  • Prednisone for maintenance immunosuppression.
  • Cyclosporine A is substituted for Tacrolimus or Azathioprine is substituted for MMF because of toxicity.
  • participants will continue their maintenance immunosuppressive regimen, but Prednisone will be held as they receive intravenous Methylprednisolone.
  • Standard of care (SOC) treatment for AMR varies based on patient-specific factors including overall performance status, previous rejection treatments, lung biopsy findings at the time of AMR diagnosis, and history of infection.
  • SOC Standard of care
  • the most common treatment combination consists of Carfilzomib, rabbit anti-thymocyte globulin (ATG), high-dose corticosteroids, and IVIG.
  • Carfilzomib 20 mg/m 2 is given on days 1, 2, 8, 9, 15, and 16, ATG (1-1.5 mg/kg) is given on days 1-5 targeting a cumulative dose of 5-7.5 mg/kg, methylprednisolone 1 mg/kg is given daily for 7-10 days with a tapering corticosteroid schedule based on clinical response, and IVIG 500-1000 mg/kg is first given on day 10 and continued monthly thereafter.
  • PLEX is not routinely used in the management of AMR because of lack of efficacy and confounding the dosing of antibody treatments including ATG and IVIG.
  • a single dose of Rituximab 375 mg/m 2 may be administered instead of ATG or for refractory disease at least 72 hours apart from the first dose of IVIG.
  • Siltuximab After enrolling in the study, subjects will receive the first dose of Siltuximab intravenously on day 1. Siltuximab will be given on the same day as the first dose of Carfilzomib and rabbit ATG. These would all be administered in the hospital. Subsequent doses may be given either during the hospitalization or in an outpatient infusion center depending on the patient's clinical status.
  • Bronchoscopy specimens are routinely tested for mycobacterial, fungal, and bacterial organisms as well as polymerase chain reaction (PCR) testing for CARV.
  • PCR polymerase chain reaction
  • positive mycobacterial, fungal, or bacterial cultures do not necessarily represent invasive disease or infection
  • patients are routinely started on appropriate antimicrobial therapy before initiating treatment for AMR to minimize the risk of invasive disease and infection after treatment for AMR.
  • Patients who have positive cultures for a mold from bronchoscopy specimens are routinely treated with Voriconazole, Posaconazole, or Isavuconazonium.
  • patients who have a positive bacterial culture are treated with appropriate antimicrobials.
  • anti-mycobacterial therapy is initiated depending on the specific organism.
  • patients who have a positive mycobacterial culture with a potentially pathogenic organism e.g., Mycobacterium abscessus, Mycobacterium avium complex
  • AMR AMR-refractory allograft dysfunction due to AMR.
  • Rescue treatments for AMR may include plasma exchange and Eculizumab, a monoclonal antibody to complement component (C5). The decision to add one of these treatments will be made by the treating lung transplant clinician. If during the course of therapy with Siltuximab a participant requires rescue treatment, subsequent doses of Siltuximab will not be given, and the patient will continue study follow-up per protocol. The treating lung transplant clinician may decide to use rescue therapy with Tocilizumab for progressive or refractory AMR. There are limited data in the literature about the use of rescue Tocilizumab after treatment with Siltuximab.
  • CRS cytokine release syndrome
  • ICANS immune effector cell associated neurotoxicity syndrome
  • rescue Tocilizumab has been given for persistent grade 1 CRS at 7 days after the first dose of Siltuximab or at 72 hours for grade 2 CRS.
  • rescue Tocilizumab may be given 4 half-lives (64 days) after the last administered dose of Siltuximab.
  • Sites will identify potential study participants from their cohorts of lung transplant recipients.
  • the research study will be explained in lay terms to each potential participant, and the potential participant will sign an informed consent form before undergoing any study procedures.
  • the investigator or study research coordinator will meet with the study candidate to review all the required elements of informed consent. After the informed consent has been signed, and the participant meets eligibility criteria, the participant will be enrolled in the study.
  • IL-6 bioavailability will be assessed by reporter gene assay (RGA) which employs an IL-6-responsive reporter cell line (DS-l/SIE-luciferase) that has been shown to have high dynamic range and specificity for IL-6 blockade in the setting of treatment with anti-IL-6 or anti-IL-6R monoclonal antibodies (80).
  • RAA reporter gene assay
  • Additional study-related testing includes baseline serum and exhaled breath condensate (EBC) IL-6 concentrations, and serial measurements of donor-derived cfDNA, DSA, and spirometry as functional biologic assessments of treatment outcome.
  • EBC IL-6 concentrations will be assessed as an exploratory metric at baseline to assess intra-pulmonary levels as serum levels may not reflect intrapulmonary production (57, 58, 60).
  • EBC IL-6 concentrations are significantly higher among cigarette smokers and patients with chronic obstructive pulmonary disease (COPD) than healthy controls but have not been measured in LT recipients (81, 82). Fasting lipids and uric acid levels may be affected by IL-6 blockade and will be measured serially.
  • COPD chronic obstructive pulmonary disease
  • the participant elects to withdraw consent from all future study activities including follow-up.
  • An SAR is any adverse event for which there is a reasonable possibility that the study drug caused the adverse event.
  • "reasonable possibility” means there is evidence to suggest a causal relationship between the drug and the adverse event.
  • An SAR implies a lesser degree of certainty about causality than adverse reaction, which means any adverse event caused by a drug.
  • An adverse event or suspected adverse reaction is considered “unexpected” if it is not listed in the most recent version of the Siltuximab Investigator Brochure (IB) or is not listed at the specificity, severity or rate of occurrence that has been observed as described in the IB; or is not consistent with the risk information described in the general investigational plan or elsewhere in the IND. "Unexpected” also refers to adverse events or suspected adverse reactions that are mentioned in the IB as occurring with a class of drugs or as anticipated from the pharmacological properties of the drug but are not specifically mentioned in the IB as occurring with the particular drug under investigation.
  • a life-threatening event an AE or SAR is considered "life-threatening" if, in the view of the investigator, its occurrence places the subject at immediate risk of death. It does not include an AE or SAR that, had it occurred in a more severe form, might have caused death,
  • Important medical events that may not result in death, be life threatening, or require hospitalization may be considered serious when, based upon appropriate medical judgment, they may jeopardize the subject and may require medical or surgical intervention to prevent one of the outcomes listed above.
  • the study site will grade the severity of adverse events experienced by participants according to the criteria set forth in NCI-CTCAE, version 5.0. This document provides a common language to describe levels of severity, to analyze and interpret data, and to articulate the clinical significance of all adverse events. Adverse events will be graded on a scale from 1 to 5 according to the following standards in the NCI-CTCAE manual:
  • a treatment-emergent adverse event is defined as an increase in grade from baseline or from the last post-baseline value that doesn’ t meet grading criteria. Changes in grade from screening to baseline will also be recorded as adverse events but are not treatment emergent. If a specific event or result from a given clinical or laboratory evaluation is not included in the NCI-CTCAE manual, then an abnormal result would be considered an adverse event if changes in therapy or monitoring are implemented because of the event/result.
  • Example 3 A Phase 1 Clinical Trial of Siltuximab for the Treatment of Antibody-Mediated Rejection after Lung Transplantation (Updated)
  • Phase 1 Clinical Trial protocol is based on the protocol set out in Example 2 but has been updated after consultation with regulatory agencies.
  • the secondary objectives are:
  • the primary endpoint is the incidence of CTCAE > grade 3 during a period of 90 days after randomization.
  • H-CRP Undetectable serum high-sensitivity C-reactive protein
  • H-CRP serum high-sensitivity C-reactive protein
  • CMV cytomegalovirus
  • Allograft survival defined as death or undergoing re-transplantation between randomization and day 180.
  • ISHLT International Society for Heart and Lung Transplantation
  • DSA Donor-specific antibodies
  • HLA human leukocyte antigens
  • MFI Mean Fluorescence Intensity
  • Serious infection Infection requiring intravenous antimicrobial therapy and hospitalization.
  • Tissue invasive CMV is defined as the presence of signs and symptoms of end-organ disease such as enteritis, colitis, pneumonitis, retinitis and CMV replication in blood.
  • the definition of opportunistic infection may include pathogenic non-tuberculous mycobacteria (NTM).
  • NTM pathogenic non-tuberculous mycobacteria
  • the placebo arm enhances the detection of safety signals by serving as a control group to compare AE rates in the Siltuximab group to.
  • the study will use safety, pharmacokinetic, pharmacodynamic, functional biological measurements, and clinical outcomes data to determine an optimal dose for a future Phase 2 clinical trial that assesses the efficacy of Siltuximab in the treatment of AMR after LT.
  • the FDA-approved dose of Siltuximab for MCD is 11 mg/kg intravenously every 3 weeks, the optimal dose for the treatment of AMR after LT is not known.
  • the role of IL-6 and its concentrations in serum and local tissues is different in different disease states. Indeed, IL-6 concentrations in serum are generally higher in MCD than in AMR.
  • Eligible subjects will be randomized on the day of enrollment through a web-based randomization system with a 1: 1 : 1 ratio to low dose Siltuximab, full dose Siltuximab, or Placebo.
  • the randomization list will be computer generated using a permuted block randomization and stratified by enrollment site. Treatment allocation will remain concealed except to each site's study pharmacist and unblinded statistician. The study staff at each site, the participants, and the clinical team will remain blinded to treatment assignment.
  • the study will enroll adult lung transplant recipients who develop clinical definite, probable, or possible AMR according to the 2016 ISHLT definition (23) and DSA to HLA. The study will not enroll children as the lung transplant programs enrolling participants do not provide care for children. Lung transplant recipients who develop AMR have a poor prognosis. IL-6 plays an important role in mediating allograft injury, and IL-6 blockade might mitigate this. The eligibility criteria for enrollment and randomization have been determined based on known adverse events associated with IL-6 blockade and Siltuximab, the package insert, and the risk of infection associated with augmented immunosuppression.
  • Investigational Agent The investigational agents for this trial are Siltuximab (Sylvant®) and Placebo (Dextrose 5% in sterile water, D5W). Details describing the study related product labeling, supply, storage, preparation, dispensing, monitoring and accountability are described below.
  • Placebo, D5W is considered an investigational agent in this study. Sites will use only sterile D5W that is produced by an FDA registered facility as designated by the presence of a National Drug Code number on product labeling. Product information including manufacturer, lot number, and expiration date will be captured in the pharmacy accountability log at each site. Placebo will be prepared with dose volume to match Siltuximab and will be stored using the standard procedures at each site's investigational pharmacy. Placebo should be prepared using appropriate aseptic techniques using the site's D5W stock. Placebo will be prepared and dispensed by an unblinded pharmacist at the site. Placebo will be labeled by the site's investigational pharmacy with the same infusion bag label used for active Siltuximab to maintain the blind.
  • Lung transplant recipients with AMR are routinely hospitalized because of allograft dysfunction and to initiate intensive immunosuppressive treatment. Depending on the clinical course and response to treatment, some patients may be discharged from the hospital to complete treatment in the outpatient infusion center. Routine clinical treatment for AMR varies based on patient-specific factors including overall performance status, previous rejection treatments, lung biopsy findings at the time of AMR diagnosis, and history of infection. The most commonly used clinical regimen at the two sites includes high-dose corticosteroids, IVIG, anti-thymocyte globulin (ATG), and Carfilzomib. ATG is not routinely used for induction immunosuppression in the immediate period after lung transplantation at the sites, but it is used to treat persistent ACR and CLAD.
  • Carfilzomib 20 mg/m 2 is given on days 1, 2, 8, 9, 15, and 16, ATG (1-1.5 mg/kg) is given on days 1-5 targeting a cumulative dose of 5-7.5 mg/kg, methylprednisolone 1 mg/kg is given daily for 7-10 days with a tapering corticosteroid schedule based on clinical response, and IVIG 500-1000 mg/kg is first given on day 10 and continued monthly thereafter.
  • PLEX is not part of the routine first-line treatment for AMR at the sites because of lack of effectiveness clearing DSA or improving the clinical course. Additionally, PLEX confounds the dosing of antibody treatments including ATG and IVIG and requires the placement of a central intravenous pheresis catheter.
  • rescue treatments for AMR may instead include PLEX, Rituximab, and Eculizumab. References in this section to "Siltuximab” should be read as "the investigational agent", in view of the blinding. Moreover, in this study, rescue Tocilizumab may be given after the end of the study (90 days) instead of 64 days after the last administered dose of Siltuximab as in Example 2.
  • Randomization will occur after the participant has signed the informed consent form.
  • IL-6 bioavailability will be assessed by reporter gene assay (RGA) which employs an IL-6- responsive reporter cell line (DS-l/SIEIuciferase) that has been shown to have high dynamic range and specificity for IL-6 blockade in the setting of treatment with anti- IL-6 or anti-IL-6R monoclonal antibodies (80).
  • RAA reporter gene assay
  • Siltuximab concentrations will be measured using a commercially available ELISA kit specifically designed to measure Siltuximab (Kribiolisa cat # KBI1630) with a minimum detection level of 12 ng/mL and an approximately 100-fold linear detection range.
  • the ELISA kit will also be independently validated with a Siltuximab standard calibration curve generated with healthy human volunteer serum. Timepoints for measuring pharmacokinetics are detailed in the schedule of events above. At every dosing visit, a pre-dose measurement and a post-dose measurement will be made; the post-dose measurement will be made 1 hour after completion of the infusion. To avoid unblinding, pharmacokinetic and HS-CRP measurements will be batched, and the results will only be made available after completion of study follow-up. Additional testing includes baseline serum and exhaled breath condensate (EBC) IL-6 concentrations, and serial measurements of donor-derived cfDNA, DSA, and spirometry as functional biologic assessments of treatment outcome.
  • EBC exhaled breath condensate
  • EBC IL-6 concentrations will be assessed as an exploratory metric at baseline to assess intrapulmonary levels as serum levels may not reflect intrapulmonary production (57, 58, 60). EBC IL-6 concentrations are significantly higher among cigarette smokers and patients with chronic obstructive pulmonary disease (COPD) than healthy controls but have not been measured in LT recipients (81, 82). Fasting lipids and uric acid levels may be affected by IL-6 blockade and will be measured serially. Tests performed for research purposes are HS-CRP, fasting lipids, uric acid, serum and EBC IL-6 concentrations, IL-6 bioavailability, and Siltuximab concentrations.
  • COPD chronic obstructive pulmonary disease
  • HS-CRP fasting lipids, and uric acid levels can be measured using the same blood draw as the CMP and do not require additional samples.
  • serum IL-6 concentrations, IL-6 bioavailability, and Siltuximab concentrations require separate blood draws; a total of 10 mL of blood will be collected for these research tests at each testing visit. Participants will have 10 mL of blood drawn for research testing at visits 0, 1, 2, 3, 4, 5, 6, 7, and 8 for a total volume of 90 mL during study follow-up. No bronchoscopies or lung biopsies will be performed for study purposes; specifically, the study does not mandate a follow-up biopsy after treatment for several reasons.
  • Example 2 Section 9.2. In this study participants may be withdrawn from the study if the participant undergoes re-transplantation.
  • a SUSAR is defined as an adverse event that is both serious and unexpected and has a reasonable possibility of a causal relationship with Siltuximab.
  • UP are defined as any event that meets all of the following criteria:
  • X* indicates 2 measurements of pharmacokinetics on days that an investigational agent dose is given: a pre-dose measurement and a postdose measurement 1 hour after completing the infusion.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

This invention relates to the use of IL-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject. In particular, this invention relates to methods of treatment of antibody mediated rejection (AMR) of a lung transplant in a subject in need thereof comprising a combination therapy of (i) administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6; and (ii) administration to the subject of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.

Description

USE OF IL-6 INHIBITORS FOR PREVENTING OR INHIBITING LUNG TRANSPLANTATION REJECTION IN A SUBJECT
CROSS-REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. Provisional Application No. 63/637,004, filed on April 22, 2024, which is hereby incorporated by reference in its entirety.
REFERENCE TO A SEQUENCE LISTING
This application contains a Sequence Listing XML, which has been submitted electronically and is hereby incorporated by reference in its entirety. Said Sequence Listing XML, created on April 16, 2025, is named P93947PC_sequence_listing_2025- 04-16 and is 8,692 bytes in size.
FIELD
This invention relates to the use of IL-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject. In particular, this invention relates to methods of treatment of antibody mediated rejection (AMR) of a lung transplant in a subject in need thereof comprising a combination therapy of (i) administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6; and (ii) administration to the subject of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
BACKGROUND
Lung transplantation (LT) is the ultimate treatment option for individuals with advanced lung disease, and approximately 2,500 people undergo lung transplantation annually in the US (1). LT improves quality of life and survival; however, long-term outcomes remain disappointing, and the median survival after LT is 6.7 years (2). Chronic lung allograft dysfunction (CLAD) is the leading cause of death beyond the first year after LT (2,3). Two phenotypes of CLAD are recognized; bronchiolitis obliterans syndrome (BOS) is characterized by an obstructive ventilatory defect without radiographic opacities, and restrictive allograft syndrome (RAS) is characterized by a restrictive ventilatory defect and upper-lobe predominant fibrotic opacities on imaging studies (4,5). A mixed phenotype with features of both BOS and RAS is also recognized. CLAD typically follows a progressive clinical course that culminates in respiratory failure and death or retransplantation.
Indeed, the median survival after the diagnosis of CLAD is approximately 2.5 years (6). Although all phenotypes are generally progressive, RAS and the mixed phenotype are typically more aggressive and result in significantly worse survival than BOS (7-9). CLAD is widely viewed as the end-result of innate and alloimmune insults that damage the allograft (10,11). These include gastroesophageal reflux disease (GERD), primary graft dysfunction (PGD), acute cellular rejection (ACR), lymphocytic bronchiolitis, donor-specific antibodies (DSA) to mismatched human leukocyte antigens (HLA), antibody-mediated rejection (AMR), and community-acquired respiratory viral (CARV) infections (12-22).
AMR is an increasingly recognized form of lung rejection, in large part because of improved awareness and the development of a standardized definition that facilitates research and communication between centres.
According to the International Society for Heart and Lung Transplantation (ISHLT) definition of AMR, the diagnosis is based on 5 criteria: DSA, abnormal lung histology, allograft dysfunction, complement component 4d (C4d) deposition, and the exclusion of other potential causes of allograft dysfunction (23). Diagnostic certainty depends on the number of criteria present; for example, a diagnosis of definite AMR is made if all 5 criteria are present, and a diagnosis of probable AMR is made if 4 of the 5 criteria are present.
The incidence of AMR has varied in previous studies depending on study design and diagnostic certainty used to identify cases. In several older single-centre retrospective cohort studies the incidence of AMR varied between 4-25% (20, 21, 24-26). In more recent multicentre prospective studies that used donor-derived cell-free DNA (cfDNA) as part of clinical monitoring, the incidence of AMR was 27% (27,28). A variety of immunosuppressive treatments focused on antibody-depletion have been used, and generally, the standard of care for AMR patients, is a multi-drug regimen including various combinations of high-dose corticosteroids, intravenous immune globulin (IVIG), Rituximab, Bortezomib or Carfilzomib, anti-thymocyte globulin (ATG), and plasma exchange (PLEX) (20, 21, 24-28). However, there have been no randomized controlled trials (RCT) and no head-to-head comparisons of different regimens to guide management. Moreover, outcomes after the diagnosis of AMR are dismal. Approximately 20% of patients die within 30 days, 50% die within 1 year, and 80% die within 2 years of the diagnosis of AMR (20, 21, 27). The majority of survivors develop CLAD early after the diagnosis of AMR. In a multicentre study, 73% developed severe CLAD, defined as >50% loss of lung function compared to baseline, within 1 year of the diagnosis of AMR (20, 21, 24, 25). Indeed, the leading causes of death after AMR are progressive CLAD and refractory AMR (20, 21, 24-27). Clearly, identifying better treatment regimens for the management of AMR is a critical unmet need for LT recipients.
All documents referred to herein are incorporated by reference in their entirety.
The listing or discussion of an apparently prior-published document in this specification should not necessarily be taken as an acknowledgement that the document is part of the state of the art or is common general knowledge.
SUMMARY OF INVENTION
An object of the invention is to provide a treatment for antibody mediated rejection (AMR) in lung transplant recipients.
In a first aspect, the present invention provides a method of treatment of antibody mediated rejection (AMR) of a lung transplant, the method comprising a combination therapy of: (i) administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6; and (ii) administration to the subject of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
In a second aspect, the present invention provides an anti-IL-6 antibody or fragment for use in treatment of an antibody mediated rejection (AMR) of a lung transplant, wherein the treatment comprises a combination therapy of administration of the antibody or fragment and administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
In a third aspect, the present invention provides a use of an anti-IL-6 antibody or fragment in treatment of antibody mediated rejection (AMR) of a lung transplant, wherein the treatment comprises a combination therapy of administration of the antibody or fragment and administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
In a fourth aspect, the present invention provides a use of an anti-IL-6 antibody or fragment for the manufacture of a medicament for treatment of antibody mediated rejection (AMR) of a lung transplant, wherein the medicament is to be administered in a combination therapy with administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
DETAILED DESCRIPTION
IL-6 is a pleiotropic cytokine that can be secreted by many cell types upon appropriate stimulation and promotes inflammatory, immune, and fibrotic responses. Different modes of IL-6 signalling exist.
In classic signalling, IL-6 binds to its receptor (IL-6R), which is devoid of signalling capacity, and the complex of IL-6 and IL6R binds to glycoprotein 130 (gpl30) which dimerizes and initiates intracellular signalling (29).
In trans-signalling, the soluble form of IL-6R (sIL-6R) binds to IL-6, and the complex binds to gpl30 inducing dimerization and intracellular signalling (29). This enables the transmission of IL-6R-initiated signals to cells that do not express the receptor thereby expanding the activation capacities of IL-6.
Both classic and trans-signalling activate signal transduction pathways leading to janus kinase (JAK)-dependent signal transducer and activator of transcription (STAT) 3 signalling as well as PI3K/AKT and RAS/MAPK pathways (30).
Trans-presentation is a third mode of IL-6 signalling where IL-6 binds to IL-6R in the cytoplasm of dendritic cells and the complex is transported to the plasma membrane where it binds to gpl30 expressed by CD4+ T-cells (31). Trans-presentation was recently discovered in mice but has not been confirmed in humans.
IL-6 expands effector and memory CD8+ T-cells by augmenting the expression of IL-2 and receptor CD122/CD25 and is essential for the differentiation of naive CD4+ T-cells towards the Thl7 phenotype (32-34). IL-17 produced by Thl7 cells promotes neutrophil proliferation and migration, endothelial cell activation, and fibroblast activation and proliferation which result in cytopathic responses implicated in acute and chronic rejection (33, 34). IL-6 was initially identified as B-cell stimulating factor 2 (BSF-2) because of its role in promoting antibody synthesis by B-cells (35-37).
Furthermore, IL-6 in conjunction with other cytokines is responsible for normal antibody production and is critical for the induction of follicular helper T-cells (Tfh) as well as the production of IL-21 which regulates immunoglobulin synthesis (38, 39). IL- 6 is also crucial for B-cell differentiation into plasmablasts and for enhancing plasmablast survival (40, 41).
Blocking IL-6 signalling effectively reduces B-cell activation, plasmablast differentiation, and antibody production (42-44). IL-6 also promotes innate immune responses. NK cells express IL-6R and are activated by IL-6 to induce endothelial cell cytotoxicity (45). IL-6 promotes fibrosis by inducing the differentiation of fibroblasts into myofibroblasts and participating in vascular smooth muscle cell and endothelial cell proliferation and activation (46). These are all important components of the pathogenesis of AMR. High concentrations of IL-6 in bronchoalveolar lavage (BAL) fluid and peripheral blood are seen with severe PGD after LT and are associated with prolonged length of stay in the intensive care unit and the hospital. However, IL-6 levels have not been consistently associated with acute cellular rejection and there have been no published results of IL-6 levels in AMR after LT. The inventors have surprisingly found that blocking IL-6 signalling may be a potential therapeutic approach for treatment of AMR in LT patients, especially when delivered with the standard of care for AMR in LT patients.
IL-6 signalling is a therapeutic target in solid organ transplantation. In a non-human primate (NHP) model of lung transplantation, all cynomolgus monkeys rejected their lung allografts in spite of conventional immunosuppression with tacrolimus, mycophenolate mofetil (MMF), and steroids, and all had DSA at the time of rejection (47). Tocilizumab (IL6R inhibitor) has also been studied for kidney (44, 49-54) and cardiac (48) AMR.
Clazakizumab (IL6 inhibitor) has been studied for AMR in kidney transplants (55, 56). In a pilot randomised controlled trial (RCT), clazakizumab attenuated the decline in GFR and depleted DSA; however, 25% of treated patients developed serious infections and 10% developed complicated diverticulitis (56). A Phase 3 RCT examining the efficacy and safety of clazakizumab for the treatment of chronic AMR after kidney transplantation is currently enrolling patients (NCT03744910).
High concentrations of IL-6 in bronchoalveolar lavage (BAL) fluid and peripheral blood are seen with severe PGD after LT and are associated with prolonged length of stay in the intensive care unit and the hospital (57-59). In contrast, IL-6 levels have not been consistently associated with ACR (60, 61). Of note, there is no association between serum and BAL fluid concentrations of IL-6 as serum concentrations do not reflect intrapulmonary production (57, 58, 60).
There have been no published results of IL-6 levels in AMR after LT. There have been no published studies of using IL-6 inhibitors, such as siltuximab, to treat AMR after LT.
The inventors have surprisingly found that from immunofluorescence staining for IL-6 in lung biopsies of patients with AMR, there is evidence of intrapulmonary IL-6 expression. These data illustrate increased IL-6 levels in serum and increased expression in lung allografts with AMR.
There are several FDA approved IL-6 inhibitors available today for treatment of rheumatoid arthritis, giant cell arteritis, systemic sclerosis-associated interstitial lung disease, cytokine release syndrome, idiopathic multicentric Castleman's disease (MCD), and neuromyelitis optica.
One example of a known IL-6 inhibitor is Siltuximab which is the only monoclonal antibody to IL-6 that is FDA-approved. In previous studies, Siltuximab has been used in patients with hematologic diseases/malignancies, solid tumors and MCD (Multicentric Castleman's Disease).
Siltuximab is a chimeric (human-murine) immunoglobulin G monoclonal antibody that binds IL-6 and is approved for the treatment of idiopathic MCD. Siltuximab prevents IL-6 from binding to both soluble and membrane bound IL-6R with high affinity and specificity resulting in deactivation of IL-6. However, there have been no published reports describing the use of Siltuximab after LT or for the treatment of AMR after any solid organ transplant. Further, there have been no published reports of use of Siltuximab in combination with routine treatment for AMR.
As discussed above, there is an unmet need for improved treatments of AMR in lung transplant patients. The present invention relies on the use of IL-6 inhibitors, such as but not limited to Siltuximab, to prevent, treat, inhibit, or block AMR or the recurrence thereof in lung transplant patients. These IL-6 inhibitors, such as Siltuximab, are to be used in combination with the routine immunosuppressive therapy (standard of care) for AMR in lung transplant recipients.
These and other aspects of the disclosure are described in detail below.
Unless indicated otherwise, all technical and scientific terms used herein will have their common meaning as understood by one of ordinary skill in the art to which this invention pertains.
It is contemplated that any method, use or treatment described herein can be implemented with respect to any other method, use or treatment described herein. All features disclosed herein in connection with any particular aspect are applicable to each of the other aspects, mutatis mutandis. In particular, all features disclosed herein in connection with the first aspect are applicable to each of the second to fourth aspects, mutatis mutandis. Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description.
The term "comprises" or "comprising" will take its usual meaning in the art, namely indicating that the component includes but is not limited to the relevant features (i.e. including, among other things). As such, the term "comprises" will include references to the component consisting essentially of (such as consisting of) the relevant features. The term "consists of" or "consisting of" will take its usual meaning in the art, namely indicating that the component includes and is limited to the relevant features.
The use of the word "a" or "an" when used in conjunction with the term "comprising" in the claims and/or the specification may mean "one," but it is also consistent with the meaning of "one or more," "at least one," and "one or more than one." The word "about" means plus or minus 5% of the stated number.
Where a numerical range is provided herein for any parameter, it is understood that all numerical subsets of that numerical range, and all the individual integer values contained therein, are provided as part of the invention. As used herein, the term "optionally" means that the subsequently described event(s) may or may not occur, and includes both event(s) which occur, and events that do not occur.
Methods of Treatment
The first aspect of the invention is a method of treatment of antibody mediated rejection (AMR) of a lung transplant in a subject in need thereof, the method comprising a combination therapy of:
(i) administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6; and
(ii) administration to the subject of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
The term "treatment", "treat" or "treating" as used herein, refers to therapeutic (curative) treatment including amelioration. Treatment also includes stopping the disease from developing or slowing further progression of the disease. For example, treatment may include preventing symptoms from worsening. The terms "prevention", "prevent" and "preventing" as used herein, refers to prophylaxis treatment i.e., action taken to prevent disease.
The terms "patient", "recipient" and "subject" are used interchangeably. By "a patient", "a subject" or "a recipient" we intend a human patient, subject or recipient. Typically the patient is an adult i.e. > 18 years old at the commencement of the treatment. Alternatively the patient may be a paediatric patient i.e. < 18 years old at the commencement of the treatment.
In a preferred embodiment of any of the aspects of the invention, the subject is a human.
In the context of this invention, "combination therapy" means that both (i) an antibody or fragment which is capable of inhibiting human IL-6; and (ii) an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6, are administered in combination as a treatment to the patient. By "in combination" we do not necessarily mean that the antibody or fragment which is capable of inhibiting human IL-6 is administered at the same time as the immunosuppressive therapy.
Antibody Mediated Rejection (AMR)
"AMR" and "antibody mediated rejection" are used interchangeably throughout this document. Antibody-mediated rejection (AMR) is a recognized cause of allograft dysfunction in lung transplant recipients. Diagnostic criteria and a working consensus definition are established by the International Society for Heart and Lung Transplantation (ISHLT) (23). AMR is a complex pathologic, serologic and clinical process.
A process of immune activation, whereby allospecific B-cells and plasma cells produce antibodies directed against donor lung antigens, is central to the concept of pulmonary AMR. The antigen-antibody complex may result in an amplified immune response, via both complement-dependent and independent pathways, which may result in lung tissue pathology and graft dysfunction to a variable degree. Complement is a multifunctional system of receptors, regulators and effector molecules that may amplify both innate and adaptive immunity contributing to the pathogenesis of AMR.
Key diagnostic criteria for AMR in lung transplant recipients may include the presence of antibodies directed toward donor human leukocyte antigens and characteristic lung histology with or without evidence of complement 4d within the graft. Exclusion of other causes of allograft dysfunction increases confidence in the diagnosis but is not essential. Pulmonary AMR may be clinical (allograft dysfunction which can be asymptomatic) or sub-clinical (normal allograft function). Both clinical and sub-clinical AMR were further sub-categorized into 3 mutually exclusive possibilities (definite, probable, and possible) by the ISHLT. These categories are based on the degree of certainty related to the presence or absence of a number of pathologic, serologic, clinical and immunologic criteria and are summarised in Table 1 (clinical) and Table 2 (subclinical).
Table 1 : Definition and Diagnostic Certainty of Clinical Pulmonary Antibody-mediated Rejection (23); + means item present; - means item absent or missing.
Table 2: Definition and Diagnostic Certainty of Sub-clinical Pulmonary Antibody- mediated Rejection (23); + means item present; - means item absent or missing.
For avoidance of doubt, there are five criteria that may be met to diagnose AMR in a lung transplant recipient. These five criteria that may be present to diagnose AMR are:
• Allograft dysfunction;
• Other potential causes of allograft dysfunction excluded;
• Abnormal lung histology;
• Lung Biopsy complement component (C4d) deposition; and
• Donor-specific antibodies (DSA).
Diagnostic certainty depends on the number of criteria present. Specifically, a diagnosis of definite clinical pulmonary AMR may be made if all five criteria are present, and a diagnosis of probable clinical pulmonary AMR may be made if any four of the five criteria are present. Specifically, a diagnosis of definite sub-clinical pulmonary AMR may be made if all three criteria (abnormal lung histology, lung biopsy C4d, and donorspecific antibodies) are present, and a diagnosis of probable sub-clinical pulmonary AMR may be made if any two of the three criteria (abnormal lung histology, lung biopsy C4d, and donor-specific antibodies) are present. Measurement and determination of each of these criteria can be achieved by any appropriate method known in the art.
In a particular embodiment of any aspect of the invention, the AMR is probable AMR or definite AMR.
AMR is increasingly recognized as a potential contributing factor to acute lung allograft dysfunction and the development of CLAD in pediatric lung transplant recipients (> 18 years of age). Although the frequency is unknown in children, AMR has clearly been documented across all pediatric age groups from infancy to early adulthood. Adult diagnostic criteria for lung allograft rejection are often applied to pediatric patients, with recent confirmation that these criteria are consistent in children.
After treatment according to the present invention, pulmonary AMR may stabilize, and/or improve. Improvement may be partial or complete. A complete response may be a return to baseline graft function if applicable, abolition of DSA titers and reversal of pathologic changes. A partial response may be an improvement in graft function if applicable, but not all parameters return to baseline. Stabilization may be defined as no further clinical deterioration.
Donor-specific anti-human leukocyte antigen (HLA) antibodies, are known to contribute to antibody-mediated rejection (AMR) in solid-organ transplantation. The best-characterized donor antigens are HLA, which are further divided, based on their structure and function, into HLA Class I and Class II. The presence of DSA at the time of transplant or detected de novo post-transplant is well described in renal transplantation, where it has been associated with compromised renal allograft survival. De novo DSA and an increase in DSA titers, perhaps via an anamnestic response, have also been associated with lung allograft dysfunction, occasionally in asymptomatic patients.
In the context of this invention, "donor-specific antibodies" is intended to mean antibodies to donor human leukocyte antigens (HLA) with a Mean Fluorescence Intensity > 1000. These can be either pre-existing or de novo. These antibodies can be measured by any appropriate method known in the art.
In a particular embodiment of any aspect of the invention, the AMR comprises the presence of donor-specific antibodies to human leukocyte antigens in the subject and/or wherein the AMR comprises allograft dysfunction. In the context of the present invention, allograft dysfunction may, for example, be considered as chronic lung allograft dysfunction.
In the context of the present invention, allograft survival may be defined as freedom of the transplant recipient from death or retransplantation.
In a particular embodiment of any aspect of the invention, the administration of the combination therapy described herein reduces one or more of the severity of allograft dysfunction, the severity of abnormal lung histology, and the amount of donor-specific antibodies in the subject. Suitably, the administration of the combination therapy described herein reduces the severity of allograft dysfunction and the amount of donorspecific antibodies in the subject, and optionally reduces the severity of abnormal lung histology. For example, the administration of the combination therapy described herein may reduce or eliminate one, two, three, four or all five criteria for AMR, such that the subject would no longer have definite AMR or probable AMR.
The skilled person would be able to determine an "abnormal lung histology" by techniques known in the art.
Lung Transplant
By "lung transplant" we mean lung transplantation, or pulmonary transplantation, which is a surgical procedure in which one (single lung transplant) or both (bilateral lung transplant) lungs are replaced by lungs from a donor. Donor lungs can be retrieved from a living or deceased donor, such as a donor that has been declared brain dead. A living donor can only donate one lung lobe.
Another type of lung transplant that is relevant to the present invention is a lobe transplant, which is a surgery in which part of a living or deceased donor's lung is removed and used to replace the recipient's diseased lung. In living donation, this procedure requires the donation of lobes from two different people, replacing a lung on each side of the recipient. Donors who have been properly screened should be able to maintain a normal quality of life despite the reduction in lung volume. In deceased lobar transplantation, one donor can provide both lobes.
A donor-recipient matching system may be used to determine an appropriate donor for a patient. For example the system administered by the United Network for Organ Sharing (UNOS) finds an appropriate match based on specific criteria, including blood type, size of organ compared with chest cavity, geographic distance between donor organ and transplant recipient, severity of the recipient's lung disease, recipient's overall health, and likelihood that the transplant will be successful.
In an embodiment of the invention, the method is initiated after the transplant of a lung from a donor. In an embodiment of the invention, the lung transplant may be a single or bilateral lung transplant. A bilateral lung transplant (BLT) is sometimes referred to as a double lung transplant or a sequential single lung transplant.
Lung transplantation may be used as a therapeutic measure of last resort for patients with end-stage lung disease who have exhausted all other available treatments without improvement. A variety of conditions may make such surgery necessary. For example, chronic obstructive pulmonary disease (COPD), including emphysema; idiopathic pulmonary fibrosis; cystic fibrosis; idiopathic (formerly known as "primary") pulmonary hypertension; alpha 1-antitrypsin deficiency; replacing previously transplanted lungs that have since failed; and other causes, including bronchiectasis and sarcoidosis. A double lung transplant may be required for example in patients with cystic fibrosis.
Some patients may require a combined heart and lung transplantation, where the patient receives a heart and two lungs. This type of transplant may be required for example where a patient has a congenital condition(s) or patients in whom a lung condition has caused significant heart disease.
IL-6 inhibitors
By "IL-6" we include any natural or synthetic protein with structural and/or functional identity to the human IL-6 protein, such as defined in UniProt Accession No. P05231, or natural variants thereof. IL-6 gene and/or amino acid sequences are disclosed in references 84-86.
By "capable of inhibiting human IL-6" we intend that any antibody or fragment that can or may be able to bind to, inhibit, block or reduce human IL-6. IL-6 exerts its biological functions via two major pathways: classic signaling and trans-signaling pathways (87). In the classic signaling pathway, IL-6 binds to the IL-6 receptor (IL- 6R) on hepatocytes and some leukocytes. The IL-6 IL-6R complex further recruits the ubiquitously expressed membrane-bound or soluble gpl30 (sgpl30), triggering the dimerization of gpl30 and intracellular signaling. In the trans-signaling pathway IL-6 interacts with soluble IL-6R (sIL-6R) to form the IL-6 sIL-6R complex, which can bind to gpl30 on any cell and initiate intracellular signaling without a requirement for the stimulated cell to express IL-6R. An antibody which is capable of inhibiting human IL- 6 must be capable of specifically binding to human IL-6, and of inhibiting its interaction with sIL-6R or IL-6R, or otherwise preventing gpl30 activation. By "capable of specifically binding", we include the ability of the antibody or antigen-binding fragment to bind at least 10-fold more strongly to the relevant polypeptide, e.g. IL-6, than to any other polypeptide; preferably at least 50-fold more strongly and more preferably at least 100-fold more strongly. Inhibitory antibodies to IL-6 can typically be divided into two groups; and the putative epitopes on the IL-6 molecule designated Site I and Site II. Site I binders prevent binding to the IL-6R or sIL-6R and thereby prevent gpl30 activation. The Site I epitope was further characterized as comprising regions of both amino terminal and carboxy terminal portions of the IL-6 molecule. Site II- binders prevent gpl30 activation and therefore may recognize a conformational epitope involved in signalling. Binding of the antibody may be measured by surface plasmon resonance, for example, by immobilizing the antibody on a chip and using recombinant human IL-6 as analyte, as described in WO 2004/039826A1. Suitable antibodies may bind IL-6 with an affinity (Kd) of at least IO-9 M, preferably at least 10’
10 M, preferably at least 10 11 or 5 x 10 11 M. Epitope mapping to identify Site I or Site
11 binders may be performed by binding to human IL-6-mutant proteins (88). Inhibition of IL-6 activity may be measured by assaying proliferation of the murine B myeloma cell line, 7TD1, in response to IL-6, as described in WO 2004/039826A1. Suitable antibodies may inhibit >50%, such as >90%, such as substantially 100% of 7TD1 cell proliferation in response to IL-6.
By "antibody" we include substantially intact antibody molecules, as well as chimeric antibodies, humanised antibodies, human antibodies (wherein at least one amino acid is mutated relative to the naturally occurring human antibodies), single chain antibodies, bi-specific antibodies, antibody heavy chains, antibody light chains, homodimers and heterodimers of antibody heavy and/or light chains, and antigen binding fragments and derivatives of the same. The term also includes antibody-like molecules which may be produced using phage-display techniques or other random selection techniques for molecules. The term also includes all classes of antibodies, including IgG, IgA, IgM, IgD, and IgE. Also included for use in the invention are antibody fragments such as Fab, F(ab')2, Fv, Fab', scFv (single-chain variable fragment), or di- scFv and other fragments thereof that retain the antigen-binding site. Similarly, the term "antibody" includes genetically engineered derivatives of antibodies such as single-chain Fv molecules (scFv) and single-domain antibodies (dAbs).
Preferred antibodies are chimeric, such as mouse-human chimeric antibodies, CDR- grafted antibodies, humanised antibodies, or human antibodies. Although the antibody may be a polyclonal antibody, it is preferred if it is a monoclonal antibody, or that the antigen-binding fragment is derived from a monoclonal antibody. Suitable monoclonal antibodies may be prepared by known techniques, for example those disclosed in "Monoclonal Antibodies; A manual of techniques" (89) and in "Monoclonal Hybridoma Antibodies: Techniques and Application" (90). The antibodies may be human antibodies in the sense that they have the amino acid sequence of human antibodies with specificity for the IL-6; however, it will be appreciated that they may be prepared using methods known in the art that do not require immunisation of humans. Suitable antibodies may be prepared from transgenic mice which contain human immunoglobulin loci, as described in "Complete humanization of the mouse immunoglobulin loci enables efficient therapeutic antibody discovery" (91).
Suitably prepared non-human antibodies can be "humanised" in known ways, for example, by inserting the CDR regions of mouse antibodies into the framework of human antibodies. Chimeric antibodies are discussed in Neuberger et al (92).
It will be appreciated by persons skilled in the art that the binding specificity of an antibody or antigen-binding fragment thereof is conferred by the presence of complementarity determining regions (CDRs) within the variable regions of the constituent heavy and light chains. As discussed below, in a particularly preferred embodiment of the antibodies and antigen-binding fragments, binding specificity for IL-6 is conferred by the presence of one or more and typically all six of the CDR amino acid sequences defined herein.
Preferably, the antibody or antigen-binding fragment comprises an antibody Fc region. It will be appreciated by the skilled person that the Fc portion may be from an IgG antibody, or from a different class of antibody (such as IgM, IgA, IgD, or IgE). For example, the Fc region may be from an IgGl, IgG2, IgG3, or IgG4 antibody. Advantageously, however, the Fc region is from an IgGl antibody. It is preferred that the antibody or antigen-binding fragment is an IgG molecule, or is an antigen-binding fragment or variant of an IgG molecule. Suitable antibodies and fragments are described in WO 2004/039826A1. Suitably, the antibody or fragment which is capable of inhibiting human IL-6 is a chimeric, humanized or CDR grafted antibody or fragment thereof. Suitably, the antibody or fragment which is capable of inhibiting human IL-6 is a chimeric, humanized or CDR grafted antibody or fragment thereof comprising a heavy chain variable region in which CDR1, CDR2, and CDR3 comprise the amino acid sequences SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO: 3, respectively; and a light-chain variable region in which CDR1, CDR2, and CDR3 comprise the amino acid sequences SEQ ID NO: 4, SEQ ID NO:, 5 and SEQ ID NO: 6, respectively, and a constant region derived from a human IgG antibody.
VH CDR1 Ser Phe Ala Met Ser (SEQ ID NO. 1)
VH CDR2 Glu He Ser Ser Gly Gly Ser Tyr Thr Tyr Tyr Pro Asp Thr Vai Thr Gly (SEQ ID NO. 2)
VH CDR3 Gly Leu Trp Gly Tyr Tyr Ala Leu Asp Tyr (SEQ ID NO. 3) VL CDR1 Ser Ala Ser Ser Ser Vai Ser Tyr Met Tyr (SEQ ID NO. 4) VL CDR2 Asp Thr Ser Asn Leu Ala Ser (SEQ ID NO. 5) VL CDR3 Gin Gin Trp Ser Gly Tyr Pro Tyr Thr (SEQ ID NO. 6)
In a preferred embodiment the antibody is siltuximab, or an antigen-binding fragment thereof. Siltuximab, also known as CNTO328 and CLLB8, with the US FDA UNII Identifier T4H8FMA7IM and the WHO ATC code L04AC11 is a chimeric (human-murine) IgGlK monoclonal antibody that binds to human IL-6. The intact molecule contains 1324 amino acid residues and is composed of two identical heavy chains (approximately 50 kDa each) and two identical light chains (approximately 24 kDa each) linked by inter-chain disulfide bonds. Siltuximab contains the antigen-binding variable region of the murine antibody, CLB-IL-6-8, and the constant region of a human IgGlK immunoglobulin.
The complete amino acid sequences of the heavy and light chains of siltuximab are shown below.
SEQ ID NO. 7 Siltuximab heavy chain amino acid sequence
EVQLVESGGKLLKPGGSLKLSCAASGFTFSS FAMSWFRQS PEKRLEWVAE I SSGGSYTYY PDTVTGRFTI SRDNAKNTLYLEMSSLRSEDTAMYYCARGLWGYYALDYWGQGTSVTVSSA STKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPE PVTVSWNSGALTSGVHTFPAVLQSSG LYSLSSVVTVPSSSLGTQTYI CNVNHKPSNTKVDKKVE PKSCDKTHTCPPCPAPELLGGP SVFLFPPKPKDTLMI SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS TYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPRE PQVYTLPPSRDEL TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGS FFLYSKLTVDKSRWQ QGNVFSCSVMHEALHNHYTQKSLSLS PGK
SEQ ID NO. 8 Siltuximab light chain amino acid sequence
QIVLIQS PAIMSAS PGEKVTMTCSASSSVSYMYWYQQKPGSS PRLLIYDTSNLASGVPVR FSGSGSGTSYSLTI SRMEAEDAATYYCQQWSGYPYTFGGGTKLE IKRTVAAPSVFI FPPS DEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTL SKADYEKHKVYACEVTHQGLSS PVTKS FNRGEC
Siltuximab and methods of preparing it, including by recombinant expression of encoding nucleic acid sequences, are described in WO 2004/039826A1.
Other suitable antibodies include olokizumab, which is a IgG4K antibody humanized from rat (93); elsilimomab (also known as B-E8), which is a mouse IgGlK monoclonal antibody (94); or the human monoclonal antibody clone 1339, which is a high-affinity fully humanized anti-IL-6 monoclonal antibody (IgGl) derived from elsilimomab (95). Further suitable antibodies include clazakizumab (formerly ALD518 and BMS-945429), which is an aglycosylated, humanized rabbit IgGl monoclonal antibody against interleukin-6 (96); sirukumab, which is a human monoclonal IgGl kappa antibody (97). Further suitable antibodies include: the MH166 antibody (98); the SK2 antibody (99); Levilimab, which is an anti IL-6 monoclonal antibody initially developed to treat rheumatoid arthritis (100); and ARGX-109, which is a preclinical stage human antibody candidate developed by arGEN-X from its SIMPLE Antibody™ platform and which is said to have outstanding neutralization potency for IL-6. Fragments of any of these antibodies may also be used.
The antibody or fragment should be prepared under sterile conditions. The appropriate volume of antibody or fragment should be withdrawn from the vials. It is recommended that the antibody solution is filtered (0.2 to 1.2 pm) before injection into the patient either by using an in-line filter during infusion or by filtering the solution with a particle filter (e.g., filter Nr. MF1830, Impromediform, Germany). The volume of the antibody is typically added to an infusion bag containing 5% dextrose. Siltuximab is available as a single-use vial containing 100 mg or 400 mg siltuximab powder for concentrate for solution for infusion, and should be stored at refrigeration temperature. The siltuximab powder is typically provided with one or more excipients, typically histidine, histidine hydrochloride monohydrate, polysorbate 80, and sucrose. After reconstitution with single-use sterile water for injection, the solution contains 20 mg siltuximab per mL. Antibodies or fragments may be formulated in other ways, as known in the art.
The dose of the antibody when administered by intravenous administration, such as by infusion, may be 11 ± 3 mg/kg patient body weight, optionally 11 mg/kg. A suitable dose of the fragment is a dose having an equivalent antagonistic effect on human IL- 6.
The combination therapy of the present invention partly comprises administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6, not human IL-6R.
Without wishing to be bound by theory, the inventors consider that inhibitors of human IL6 will be advantageous over inhibitors of IL-6R for treating AMR in lung transplant recipients for many reasons.
For example, treatment with monoclonal antibodies to IL-6R may result in significant increases in IL-6 and IL-6R levels (44, 65). Although this may not cause deleterious effects while treatment is ongoing, there is a theoretical possibility that a rebound IL- 6 effect may be seen when treatment is discontinued. Indeed, the occurrence of allograft loss in 4 kidney recipients with chronic AMR who stopped Tocilizumab supports this possibility (50, 56). Furthermore, the inventors consider that considerably lower doses of monoclonal antibodies to IL-6 may be used compared to those to IL-6R because soluble IL-6R is present at high concentrations compared to IL-6, and all soluble IL-6R have to be saturated with a therapeutic antibody to exert its pharmacodynamic effect (66). In addition, IL-6R binds not only IL-6 but also IL-27p28, so the blockade of IL-6R may affect other signaling pathways (67). Turning to specific inhibitors, Siltuximab (IL6 inhibitor) has a higher binding affinity for IL-6 than Tocilizumab (IL-6R inhibitor) has for IL-6R (68).
In a preferred embodiment of any of the aspects of the invention, the antibody or fragment which is capable of inhibiting human IL-6 is selected from siltuximab, olokizumab, elsilimomab, mAb 1339, clazakizumab, sirukumab, levilimab and ARGX- 109.
Siltuximab In a particularly preferred embodiment of any of the aspects of the invention, the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab.
In previous studies, Siltuximab has been used in patients with hematologic diseases/malignancies, solid tumors and MCD. In Phase 1 studies that included patients with advanced solid tumors, refractory multiple myeloma, or MCD, common grade > 3 adverse events (AE) included leukopenia, neutropenia, thrombocytopenia, hypercholesterolemia, anemia, fatigue, infections, and abnormal liver enzymes (62, 69-71). In Phase 2 studies in MCD, Siltuximab has generally been well-tolerated during long-term follow-up, with a similar pattern of grade > 3 AEs as in Phase 1 studies (72- 74). Siltuximab's safety data are consistent with those of other IL-6/IL-6R monoclonal antibodies (64). Serious infections are the most common serious adverse events (SAE) in clinical trials and post-marketing surveillance studies; other SAE include GI tract perforation, hepatitis, and pancreatitis (64).
Siltuximab has not previously been used in combination with routine treatment for AMR.
Siltuximab dosing studies for indications other than AMR have examined different doses ranging between 3 mg/kg to 15 mg/kg with most doses given every 2 or 3 weeks in patients with hematologic diseases/malignancies, solid tumors, or Multicentric Castleman's Disease (62, 70, 71, 75,76). Measuring IL-6 concentrations accurately during treatment with Siltuximab is not feasible because Siltuximab-neutralized IL-6 complexes distort quantification methods (76). Thus, levels of C-reactive protein (CRP) are used as a surrogate for IL-6 activity because IL-6 is the primary factor that drives CRP production by hepatocytes (76, 77). In these studies, doses of 11 mg/kg or 15 mg/kg every 3 weeks resulted in the most pronounced suppression of CRP, and the half-life of Siltuximab was approximately 21 days (62, 70, 71, 76). Consequently, the currently approved dose of Siltuximab for MCD is 11 mg/kg every 3 weeks. However, because suppression of CRP to levels below 1 mg/L may not be seen until after the second dose, weekly dosing for 4 weeks is recommended for patients with severe MCD (78). No apparent differences in pharmacokinetics were observed across different disease states, and serum concentrations of Siltuximab reached steady state by the sixth infusion. The mean terminal half-life of the 11 mg/kg dose is 20.6 days (range: 14.2-29.7 days).
Immunosuppressive Therapy By "immunosuppressive therapy" we include the standard, routine, common immunosuppressive therapies which are given to transplant recipients before, after or during the transplant procedure to suppress their immune system and reduce the risk of the recipient's body rejecting the donor lung(s) (transplant). The purpose is to stop an unwanted immune response that would damage survival of the transplanted tissue and/or of the subject. Some transplant recipients may be on a life-long immunosuppressive treatment regimen after the transplant. Some patients may be able to reduce or stop taking certain immunosuppressive therapies (e.g., corticosteroids) after the lung transplant, which may reduce the risk of the side effects and complications associated with the immunosuppressive therapies. This is particularly because continuation of immunosuppressive therapies may result in immunodeficiency, by which the subject may be susceptible to opportunistic infection and/or increase the likelihood of cancer due to reduced surveillance of damaged cells by the immune system.
Some common immunosuppressive therapy options administered to patients after a lung transplant include glucocorticoids (corticosteroids), monoclonal antibodies (mAbs), nucleotide blocking agents, calcineurin inhibitors, and mTOR inhibitors. Although monoclonal antibodies are known immunosuppressants, antibodies or fragments which are capable of inhibiting human IL-6 are not known for use in treatment of AMR in lung transplant patients. Suitable immunosuppressive therapy options are known to the person skilled in the art.
The immunosuppressive therapy may comprise a multi-drug regimen. For example, the combination therapy may comprise two or more of the immunosuppressive therapy treatments described herein.
In particular, the immunosuppressive therapy may comprise administering one or more of a baseline immunosuppression maintenance therapy, and/or a standard of care treatment for AMR.
Immunosuppressive therapies may be categorised as induction agents, maintenance therapy, or rescue therapy (i.e. to treat rejection when observed). Induction agents are administered at the time of transplant and are generally more powerful immunosuppressive agents; for this reason they are given for a short acute period. Maintenance therapy comprises medications used over a longer period. Rescue therapy is given when clinical signs of rejection require additional action, such as when the transplant rejection becomes refractory to other medications. In the context of this invention, "baseline immunosuppression maintenance therapy" is defined as immunosuppressive therapy that is categorised as maintenance therapy and used as a form of therapy to be administered chronically to ensure and improve tolerance and continued acceptance of the recipient organ or graft. It typically is a combination of agents and includes one or more of calcineurin inhibitors (cyclosporine and tacrolimus), mammalian target of rapamycin (mTOR) inhibitors (sirolimus and everolimus), antiproliferative agents (azathioprine and mycophenolic acid), costimulatory blockers (belatacept), and corticosteroids.
In general, lung transplant recipients may be treated with a combination of Tacrolimus, Mycophenolate Mofetil (MMF), and Prednisone (or methylprednisolone) for maintenance immunosuppression. In some cases, Cyclosporine A is substituted for Tacrolimus or Azathioprine is substituted for MMF because of toxicity. In accordance with the present invention, patients may be administered a routine maintenance immunosuppression therapy as described herein.
In particular, the baseline immunosuppression maintenance therapy may comprise one or more of tacrolimus, cyclosporine A, Mycophenolate Mofetil (MMF), azathioprine, and prednisone. Suitably, the baseline immunosuppression maintenance therapy may comprise:
(a) tacrolimus, or cyclosporine A;
(b) Mycophenolate Mofetil (MMF), or azathioprine; and
(c) optionally prednisone.
In the context of the present invention, the standard of care treatment for AMR is defined as a routine or standard treatment for pulmonary AMR that is commonly or generally administered to a patient before, during or after receiving a lung transplant. For example, this may include preventative immunosuppressive treatment administered before, during or after transplantation of one or more lung(s) or lobe(s) thereof, to prevent AMR developing, and/or routine immunosuppressive treatment administered before, during or after transplantation to ameliorate, stabilise, improve, or (partially) treat AMR in a lung transplant recipient.
The standard of care treatment for AMR may comprise one or more of a corticosteroid, intravenous immune globulin (IVIG), Rituximab, Bortezomib, Carfilzomib, antithymocyte globulin (ATG), plasma exchange (PLEX), tacrolimus, cyclosporine A, Mycophenolate Mofetil (MMF), azathioprine, prednisone, and methylprednisolone, and optionally acetaminophen (paracetamol) and/or diphenhydramine.
The corticosteroid may be selected from one or more of prednisolone, prednisone, methylprednisolone, cortisone, dexamethasone, betamethasone and hydrocortisone. These are all commercially available corticosteroids.
In particular, the immunosuppressive therapy (standard of care for AMR) may comprise acetaminophen, diphenhydramine, and methylprednisolone. In such cases, the acetaminophen, diphenhydramine, and methylprednisolone may be administered 30- 60 minutes prior to an or each administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6. For example, the antibody or fragment which is capable of inhibiting human IL-6 may be siltuximab.
In particular, the administration of the immunosuppressive therapy may comprise the administration of acetaminophen 650 or 1000 mg by mouth (oral administration e.g, by tablet or capsule), diphenhydramine 25 or 50 mg by mouth (oral administration e.g, by tablet or capsule), and methylprednisolone 60 mg intravenously. In this case, antibody or fragment which is capable of inhibiting human IL-6 is preferably Siltuximab. Further, the acetaminophen, diphenhydramine, and methylprednisolone may be administered 30-60 minutes prior to an or each administration of the antibody of fragment (e.g., Siltuximab). The acetaminophen, diphenhydramine, and methylprednisolone do not necessarily need to be administered with every dose of the antibody or fragment but they may be administered with each dose of antibody or fragment (e.g., Siltuximab) if required.
The skilled person would be able to design a suitable treatment plan of the immunosuppressive therapy according to the needs of the patient. The treatment plan may be revised during the treatment (e.g. between doses of the antibody or fragment) as the patient requires (for example in reaction to the patient's toleration of a particular dose or type of antibody or fragment, or to take into account the perceived efficacy of treating AMR in a particular patient a particular dose or type of antibody or fragment).
Administration of the Combination Therapy
The antibody or fragment which is capable of inhibiting human IL-6 may be administered via the same or a different administration route as the immunosuppressive therapy. In one embodiment, the antibody or fragment which is capable of inhibiting human IL-6 may be prepared e.g. for parenteral administration e.g., subcutaneous, intramuscular, intravenous, intra-dermal, intra-articular or periarticular administration, particularly in the form of liquid solutions or suspensions; or for inhalation to the lungs e.g. pulmonary administration, particularly in the form of solutions, suspensions including nanosuspensions for nebulisation, or suspension or solution pressurised or non-pressurised aerosols. In such embodiments, the immunosuppressive therapy may be prepared for parenteral administration e.g., subcutaneous, intramuscular, intravenous, intra-dermal, intra-articular or peri-articular administration, particularly in the form of liquid solutions or suspensions; for oral administration, particularly in the form of tablets, capsules, powder, granules, solid dispersions or in the form of liquid solutions or suspensions including nanosuspensions; for inhalation to the lungs or nose e.g. pulmonary or intranasal administration, particularly in the form of dry powders, solutions, suspensions including nanosuspensions for nebulisation, nasal sprays or drops comprising solutions or suspensions or suspension or solution pressurised or non-pressurised aerosols; for topical or transdermal administration e.g. as creams, sprays, foams, gels, ointments, liquids, patches; for mucosal administration e.g. to buccal, sublingual or vaginal mucosa, and for rectal administration e.g. in the form of a foam or suppository.
The antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy may be administered by inhalation. An advantage of inhaled medications is their direct delivery to the area of rich blood supply in comparison to many medications taken by oral route. Thus, the absorption is very rapid as the alveoli have an enormous surface area and rich blood supply and first pass metabolism is bypassed.
The antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy may conveniently be administered in unit dosage form and may be prepared by any of the methods well-known in the pharmaceutical art. The antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy may also conveniently be administered in multiple unit dosage form.
The present invention also provides an inhalation device containing the antibody or fragment which is capable of inhibiting human IL-6, and/or the immunosuppressive therapy of the present invention. Typically said device is a metered dose inhaler (MDI), which contains a pharmaceutically acceptable chemical propellant to push the medication out of the inhaler. The immunosuppressive therapy may be administered by intranasal administration. The nasal cavity's highly permeable tissue is very receptive to medication and absorbs it quickly and efficiently. Nasal drug delivery is less painful and invasive than injections, generating less anxiety among patients. By this method absorption is very rapid and first pass metabolism is usually bypassed, thus reducing inter-patient variability.
The immunosuppressive therapy may be administered by transdermal administration. For topical delivery, transdermal and transmucosal patches, creams, ointments, jellies, solutions or suspensions may be employed.
The immunosuppressive therapy may be administered by sublingual administration.
In a preferred embodiment of any of the aspects of the invention, the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is by intravenous administration, optionally by infusion, optionally wherein the infusion is over the course of one hour.
The present disclosure provides pharmaceutical compositions comprising IL-6 inhibitors and other pharmaceutical compositions for administering immunosuppressive therapy. Such compositions may comprise a prophylactically or therapeutically effective amount of the active drug (the IL-6 inhibitor and/or a drug required by the immunosuppressive therapy), and a pharmaceutically acceptable carrier. In a specific embodiment, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" refers to a diluent, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a particular carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Other suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The various compositions in the context of the therapies and administrations described herein, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. Oral formulations can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical agents are described in "Remington's Pharmaceutical Sciences." Such compositions will contain a prophylactically or therapeutically effective amount of the agent, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration, which can be oral, intravenous, intraarterial, intrabuccal, intranasal, nebulized, bronchial inhalation, intra-rectal, vaginal, topical or delivered by mechanical ventilation.
Pharmaceutically acceptable salts include the acid salts and those which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups may also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
Generally, the ingredients of compositions of the disclosure are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water-free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
As the immunosuppressive therapy is intended to be a routine (standard) immunosuppressive therapy generally administered to lung transplant recipients, a person skilled in the art would be aware of the appropriate administration routes for different immunosuppressive therapies. For example, the administration route may vary inter alia according to patient characteristics, dosage amount(s) and/or frequency, and/or the type of immunosuppressive therapy. Further, when the immunosuppressive therapy is a multi-drug regimen, the administration route (technique) may be different across different immunosuppressive therapies within the multi-drug regimen.
The antibody or fragment which is capable of inhibiting human IL-6 may be administered according to a dosage regimen. For instance, the administration may include one or more (including two or more, three or more, four or more) doses of the antibody or fragment. For example, the administration may comprise one, two, three, four, five, six, seven, eight, nine or more doses to a subject of the antibody or fragment which is capable of inhibiting human IL-6. Preferably, the antibody or fragment which is capable of inhibiting human IL-6 is administered as 2 or more doses. For example, the antibody or fragment which is capable of inhibiting human IL-6 may be administered as 2 or 3 doses. For example, the antibody or fragment which is capable of inhibiting human IL-6 may be administered as 2 doses. For example, the antibody or fragment which is capable of inhibiting human IL-6 may be administered as 3 doses.
The dose of the antibody or fragment is determined according to the weight in kg of the patient. Where a dose is given in the units of "mg/kg", this refers to mg of antibody or fragment or other drug per kg of body weight of the patient. An antibody fragment is to be administered at an equivalent fragment dose having an equivalent antagonistic effect on human IL-6 to the whole antibody from which the fragment is derived. The equivalent fragment dose may be calculated according to the fragment molecular weight compared to the molecular weight of the whole antibody, also referred to as parent antibody. For example, if a given antibody has a molecular weight of 150 kD, and a Fab fragment has a molecular weight of 50 kD, then a fragment dose that is one third of the antibody dose should provide an equivalent antagonistic effect on human IL-6. Thus, if the antibody dose was 12 mg/kg, then the equivalent fragment dose for the Fab fragment would be 4 mg/kg. The equivalent antagonistic effect on human IL-
6 may also be determined according to the amount of human IL-6 that the fragment can specifically bind to, compared to the amount of human IL-6 that the parent antibody can specifically bind to. These amounts may be determined by various assays, including ELISA.
In a particular embodiment of any of the aspects of the invention, the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is in a dose of 1 mg/kg to 15 mg/kg, such as 1 mg/kg, 1.5 mg/kg, 2 mg/kg, 2.5 mg/kg, 3 mg/kg, 3.5 mg/kg, 4 mg/kg, 4.5 mg/kg, 5 mg/kg, 5.5 mg/kg, 6 mg/kg, 6.5 mg/kg,
7 mg/kg, 7.5 mg/kg, 8 mg/kg, 8.5 mg/kg, 9 mg/kg, 9.5 mg/kg, 10 mg/kg, 10.5 mg/kg, 11 mg/kg, 11.5 mg/kg, 12 mg/kg, 12.5 mg/kg, 13 mg/kg, 13.5 mg/kg, 14 mg/kg, 14.5 mg/kg or 15 mg/kg. For example, the dose may be 5.5 mg/kg or 11 mg/kg. Preferably, the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab. More preferably, the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab, and is administered in a dose of 5.5 mg/kg or 11 mg/kg.
Particular dosage regimens of the antibody or fragment which is capable of inhibiting human IL-6 include:
• administering to a subject on day 1 of treatment a first dose of the antibody or fragment (preferably siltuximab) (i.e., a one dose treatment); or
• administering to a subject on day 1 of treatment a first dose of the antibody or fragment (preferably siltuximab) and administering to a subject on day 8 a second dose of the antibody or fragment (preferably siltuximab) (i.e., a two dose treatment);
• administering to a subject on day 1 of treatment a first dose of the antibody or fragment (preferably siltuximab) and administering to a subject on day 22±3 (particularly day 22) a second dose of the antibody or fragment (preferably siltuximab) (i.e., a two dose treatment); and
• administering to a subject on day 1 of treatment a first dose of the antibody or fragment (preferably siltuximab), administering to a subject on day 8 a second dose of the antibody or fragment (preferably siltuximab), and administering to a subject on day 22±3 (particularly day 22) a third dose of the antibody or fragment (preferably siltuximab) (i.e., a three dose treatment).
In any of the above dosage regimens, the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is in each dose may be from 1 mg/kg to 15 mg/kg. Suitably, each dose may be 5.5 mg/kg or 11 mg/kg.
Each dose administered in the same dosage regimen does not have to be of the same dosage amount. For example, a subject may be administered a first dose of 11 mg/kg of the antibody or fragment (preferably siltuximab) on day 1 of treatment, and then administered a second dose of 5.5 mg/kg of the antibody or fragment (preferably siltuximab) on day 8 of treatment.
In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 11 mg/kg on day 8 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 11 mg/kg on day 22±3 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 11 mg/kg on day 22 of treatment.
In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, a second dose of siltuximab is administered at 11 mg/kg on day 8 of treatment, and a third dose of siltuximab is administered at 11 mg/kg on day 22±3 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, a second dose of siltuximab is administered at 11 mg/kg on day 8 of treatment, and a third dose of siltuximab is administered at 11 mg/kg on day 22 of treatment.
In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 22±3 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22±3 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment.
In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 22±3 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22±3 of treatment. In a particular embodiment of any aspect of the invention, a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment.
Further, the subject may be administered a different antibody or fragment which is capable of inhibiting human IL-6 as part of the same dosing regime. For example, the first dose may comprise siltuximab, and the second dose may comprise a different antibody or fragment which is capable of inhibiting human IL-6, such as one or more IL-6 inhibitors selected from olokizumab, elsilimomab, mAb 1339, clazakizumab, sirukumab, levilimab, SK2, MH166, and ARGX-109. A third dose may comprise the same antibody or fragment as the first and/or second dose, or a different antibody or fragment as the first and/or second dose.
Alternatively, the same antibody or fragment which is capable of inhibiting human IL- 6 may be used throughout the treatment (e.g., for each dose). Preferably, each dose comprises or consists of siltuximab.
The treatment of the present invention is a combination therapy of (i) an antibody or fragment which is capable of inhibiting human IL-6; and (ii) an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6. In particular, the immunosuppressive therapy does not comprise any of siltuximab, olokizumab, elsilimomab, mAb 1339, clazakizumab, sirukumab, levilimab, SK2, MH166, and ARGX- 109. The immunosuppressive therapy may comprise IL6R inhibitors such as Tocilizumab. However, in most embodiments it is envisioned that the only antibody inhibitor of IL-6 signalling will be the antibody or fragment which is capable of inhibiting human IL-6. Hence, in such embodiments, the immunosuppressive therapy would not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6 or human IL-6R. Moreover, it will be appreciated that in most embodiments the only inhibitor of IL-6 signaling will be the antibody or fragment which is capable of inhibiting human IL-6. Hence, in such embodiments, the immunosuppressive therapy would not comprise any drug which is capable of inhibiting human IL-6 or human IL-6R.
In an embodiment, the combination therapy may comprise (i) an antibody or fragment which is capable of inhibiting human IL-6; and (ii) an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise the antibody or fragment which is capable of inhibiting human IL-6 from (i). For example, if the antibody or fragment which is capable of inhibiting human IL-6 of (i) is Siltuximab, the immunosuppressive therapy of (ii) does comprise not Siltuximab.
The administration of (i) an antibody or fragment which is capable of inhibiting human IL-6; may occur before, after, at substantially the same time as, or during the administration of (ii) an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6. In the combination therapy of the invention, each or a dose of (i) an antibody or fragment which is capable of inhibiting human IL-6 may be administered with or without a dose of (ii) immunosuppressive therapy, as long as a dose of (ii) immunosuppressive therapy is administered at least once during the treatment.
In a particular embodiment of any aspect of the invention, the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is given in a first treatment dose initiated within the period 24 hours before or after initiation of the immunosuppressive therapy. The administration of the first treatment dose of the antibody of fragment may be initiated within the period 2 hours before or after initiation of the immunosuppressive therapy. The administration of the first treatment dose of the antibody of fragment may be initiated within the period one hour before or after initiation of the immunosuppressive therapy. The administration of the first treatment dose of the antibody of fragment may be initiated substantially at the same time as initiation of the immunosuppressive therapy.
In a preferred embodiment, the immunosuppressive therapy is administered 30 to 60 minutes prior to an or each administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6. Preferably, the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab.
This means that, in such an embodiment, the immunosuppressive therapy is administered 30 to 60 minutes prior to one or more doses (particularly the first dose, or the only dose if there is only one dose administered) of the antibody or fragment (preferably Siltuximab). If the treatment comprises more than one dose of the antibody or fragment which is capable of inhibiting human IL6, the immunosuppressive therapy for example may be administered 30 to 60 minutes prior to every dose, only the first dose, only the last dose, only the second dose, only one dose, only one or two doses out of a three dose treatment, every other dose, every second dose, or every third dose and so on, of the antibody or fragment (for example, Siltuximab).
The administration of the combination therapy may further comprise administration to the subject of a prophylactic medication, wherein the prophylactic medication is an antiviral and/or antimicrobial medication. The prophylactic medication may be one or more of an antiviral medication, or an antimicrobial medication. The person skilled in the art will appreciate that any suitable antiviral or antimicrobial medication may be administered prophylactically, i.e. to prevent possible viral or microbial infection. By microbial infection we include bacterial, mycobacterial and fungal infections, such that an antimicrobial medication may be an antibiotic agent against bacteria or mycobacteria, or an antifungal agent against fungi (particularly yeasts or molds). In particular, the prophylactic medication may comprise one or more of Valganciclovir, Trimethoprim-Sulfamethoxazole, Voriconazole, Posaconazole, and Isavuconazonium.
Such prophylactic medications are considered standard treatments for lung transplant recipients (i.e., these medications are part of the standard of care), and the skilled person would be able to determine the appropriate dosage, medication, and prophylactic treatment regimen. These prophylactic medications are known and commercially available.
Participants at risk for cytomegalovirus (i.e., seronegative recipients of seropositive donors or seropositive recipients) may be treated with Valganciclovir prophylactically after treatment for AMR. Similarly, all lung transplant recipients may be maintained on prophylaxis for Pneumocystis jirovecii with either Trimethoprim-Sulfamethoxazole or an alternative drug if allergic to sulfa.
All patients being evaluated for AMR may undergo bronchoscopy with lung biopsies, bronchial washings, and/or bronchoalveolar lavage as part of the routine clinical evaluation. Bronchoscopy specimens may be routinely tested for mycobacterial, fungal, and bacterial organisms as well as polymerase chain reaction (PCR) testing for CARV. Although positive mycobacterial, fungal, or bacterial cultures do not necessarily represent invasive disease or infection, patients may be routinely started on appropriate antimicrobial therapy before initiating treatment for AMR to minimize the risk of invasive disease and infection after treatment for AMR.
Patients who have positive cultures for a mold from bronchoscopy specimens may be routinely treated with Voriconazole, Posaconazole, or Isavuconazonium. Patients who have a positive bacterial culture may be treated with appropriate antimicrobials. For patients who have a positive mycobacterial culture, anti-mycobacterial therapy may be initiated depending on the specific organism.
DESCRIPTION OF THE FIGURES
The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
Figure 1 shows (A) serum IL-6 concentrations in a cohort of lung transplant patients with AMR compared to healthy non-transplant individuals, and (B) immunofluorescence staining for IL-6 in lung biopsies of patients with AMR.
Figure 2 shows the traditional 3+3 Phase 1 clinical trial design used in Example 2.
Figure 3 shows the dosing and follow-up timelines for Cohorts A-D in Example 2.
EXAMPLES
Example 1: Measurements of IL-6 in patients with AMR
Using an ELISA method, serum IL- 6 concentrations were measured in a cohort of LT recipients with AMR (n = 15), which revealed a median concentration of 1.94 (range: 0.91-4.26) pg/mL compared to healthy non-transplant individuals who had a median concentration of 0.56 (range: 0.06-1.83) pg/mL (Figure 1A, p < 0.001).
Studies in other disease states (e.g., multiple myeloma, non-Hodgkin's lymphoma) have shown wide interpatient variability in baseline serum IL-6 concentrations, and baseline serum concentrations were not predictive of clinical response to IL-6 blockade because serum concentrations may not reflect local production or concentrations (62).
Hence, localised detection is important. Immunofluorescence staining for IL-6 in lung biopsies of patients with AMR demonstrated evidence of intrapulmonary IL-6 expression (Figure IB).
These preliminary data illustrate increased IL-6 levels in serum and increased expression in lung allografts with AMR. Example 2: A Phase 1 Clinical Trial of Siltuximab for the Treatment of Antibody-Mediated Rejection after Lung Transplantation
This is a Phase 1, prospective, multicenter, open-label clinical trial in which 18 lung transplant recipients will be treated with escalating doses of Siltuximab in addition to standard of care therapy for antibody-mediated rejection after lung transplantation.
1. Protocol Synopsis
1.1. Primary Objective
The primary objective of the study is to assess the safety and tolerability of Siltuximab added to routine immunosuppressive treatment for antibody-mediated rejection after lung transplantation.
1.2. Secondary Objectives
The secondary objectives are:
1. Assess pharmacodynamics,
2. Determine the optimal dose for a subsequent trial, and
3. Assess functional biological measurements and clinical outcomes feasible for use in a subsequent trial that examines efficacy.
1.3. Primary Endpoint
The primary endpoint is safety and tolerability which will be assessed based on the development of dose limiting toxicities (DLT), adverse events (AE), and serious adverse events (SAE).
1.4. Secondary Endpoints
The following secondary endpoints will be assessed between enrolment and day 90:
1. Undetectable serum C-reactive protein (CRP) (< 10 mg/L),
2. Clearance of donor-specific antibodies (DSA),
3. Hyperuricemia (> 7 mg/dL),
4. Hyperlipidemia requiring the initiation or dose increase of medical therapy,
5. Confirmed bacterial, cytomegalovirus (CMV), mold, mycobacterial, or community-acquired respiratory viral infection (each assessed independently),
6. Development of probable chronic lung allograft dysfunction (CLAD) according to the 2019 ISHLT criteria (4, 5),
7. Allograft survival defined as death or undergoing re-transplantation.
1.5. Exploratory Endpoints The following exploratory endpoints will be assessed between enrolment and day 90:
1. Donor-derived cell-free-DNA < 1%,
2. Serum IL-6 levels at enrolment only (before administration of the first dose of Siltuximab),
3. Exhaled breath condensate IL-6 levels at enrolment,
4. Serial measurements of IL-6 bioavailability by reporter gene assay (RGA).
1.6. Inclusion Criteria
Individuals who meet all the following criteria are eligible for enrolment as study participants:
1. 18 years of age or older,
2. Single or bilateral lung transplant recipient,
3. New diagnosis of definite or probable antibody-mediated rejection according to the 2016 International Society for Heart and Lung Transplantation (ISHLT) definition (23),
4. Donor-specific antibodies (DSA) to human leukocyte antigens (HLA),
5. Able to understand the purpose of the study and willing to participate and sign informed consent.
1.7. Exclusion Criteria
Individuals who meet any of these criteria are not eligible for enrolment as study participants:
1. Pregnant or breast feeding,
2. History of lymphoma or hematologic malignancy,
3. Treatment with IL-6 signaling blockade with 6 months of enrolment,
4. Cancer other than non-melanoma skin cancer with disease-free period < 3 years,
5. Positive respiratory virus PCR detected within 7 days of enrolment,
6. Active cytomegalovirus infection within 7 days of enrolment,
7. Positive respiratory culture for Mycobacterium tuberculosis, Mycobacterium abscessus, Mycobacterium chelonae, or Mycobacterium avium complex within 4 weeks of enrolment,
8. Absolute neutrophil count (ANC) < 1,000 cells/mm3 at enrolment,
9. Platelet count < 75,000 cells/mm3 at enrolment,
10. Hemoglobin > 17 g/dL at enrolment,
11. ALT or AST > 2.5 times upper limit of normal at enrolment,
12. Total bilirubin > 2.5 times upper limit of normal at enrolment,
13. History of gastrointestinal tract perforation, 14. Diagnosis of diverticulitis within 4 weeks of enrolment,
15. Inability or unwillingness to give written informed consent or comply with the study protocol,
16. Any condition that in the opinion of the site investigator introduces undue risk by participating in this study or impacts the quality or interpretation of the study results.
1.8. Study Stopping Rules
The principal investigator team, the Data and Safety Monitoring Board (DSMB), and NHLBI will review safety data on an ongoing basis. If a safety concern arises, enrollment and randomization of participants in the trial will be suspended pending DSMB review. Subjects already enrolled will continue to be treated per protocol.
The criteria described below provide additional guidance for suspending the trial pending DSMB review based on the occurrence of selected adverse events. Selected adverse events of concern and their thresholds in this trial are:
1. Any occurrence of gastrointestinal tract perforation,
2. Death in greater than 20% of enrolled subjects,
3. Opportunistic infections in greater than 25% of enrolled subjects.
2. Study Definitions
Allergic reaction: An allergic reaction is defined as a disorder characterized by an adverse local or general response from exposure to an allergen. Grade 3 allergic reaction is one that results in bronchospasm, requires hospitalization, or intravenous intervention.
Allograft survival: Freedom from death or re-transplantation.
Antibody-Mediated Rejection: Antibody-mediated rejection will be defined based on the
2016 ISHLT definition. Only cases of definite or probable AMR with donor-specific antibodies (DSA) to human leukocyte antigens (HLA) will be considered as AMR in the study.
Bronchoalveolar lavage (BAL): Bronchoalveolar lavage is medical procedure performed during bronchoscopy in which a small scope is inserted into a patient's nose or mouth and passed into the lungs. Once in the lungs, sterile saline is instilled, coating the lining the lungs, and then quickly aspirated out of the lungs.
Clearance of donor-specific antibodies (DSA): Clearance of donor-specific antibodies (DSA) is defined as all DSA being below the level of detection (Mean Fluorescence Intensity < 1000). Death as a DLT: Death is defined as a Dose Limiting Toxicity if the CEAC adjudicates that death was due to infection.
Donor-specific antibodies (DSA): Donor-specific antibodies are antibodies to donor human leukocyte antigens (HLA) with a Mean Fluorescence Intensity > 1000. These can be either pre-existing or de novo.
Hepatitis as a DLT: Hepatitis is defined as a Dose Limiting Toxicity if the AST or ALT > 5 times the upper limit of normal or if the total bilirubin > 3 times the upper limit of normal.
Hyperlipidemia as a DLT: Hyperlipidemia is defined as a Dose Limiting Toxicity if it results in pancreatitis, requires hospitalization, or prolongs a hospitalization.
Hypersensitivity reaction: A hypersensitivity reaction is defined as a Dose Limiting Toxicity if this meets the definition of a Grade > 3 allergic reaction.
Infection as a DLT: Infections including CMV, fungemia, and respiratory tract infections are defined as Dose Limiting Toxicities if they require hospitalization or prolong a hospitalization.
Neutropenia as a DLT: Neutropenia is defined as a Dose Limiting Toxicity if the absolute neutrophil count < 500 cells/mm3 for at least 3 days.
Opportunistic infection: Opportunistic infections are infections that occur more often or are more severe in immunocompromised patients. These include tissue invasive cytomegalovirus, varicella zoster, tuberculosis, Pneumocystis jirovecii, nocardia, endemic mycoses, and toxoplasmosis.
Probable CLAD: Probable CLAD is defined as a >20% decline in FEVi compared to the baseline value on 2 measurements at least 3 weeks apart and after exclusion or adequate treatment of potential secondary causes of allograft dysfunction (e.g., respiratory infection, acute cellular rejection).
Pruritis as a DLT: Pruritis is defined as a Dose Limiting Toxicity if it is widespread and constant, limits self-care, or requires systemic steroids.
Rash as a DLT: Rash is defined as a Dose Limiting Toxicity if it covers > 30% of the body surface area with moderate or severe symptoms limiting self-care.
Thrombocytopenia as a DLT: Thrombocytopenia is defined as a Dose Limiting Toxicity if the platelet count < 50,000 cells/mm3 for at least 3 days.
3. Study Hypothesis
We hypothesize that the addition of Siltuximab to standard of care immunosuppressive therapy for AMR is sufficiently safe and clinically tolerable to proceed with a subsequent Phase 2 clinical trial that assesses efficacy. To test this hypothesis, we will conduct a Phase 1 clinical trial of 18 adult lung transplant recipients with AMR. 4. Study Design
4.1. Description of study design
This is a two-center, prospective, open-label, Phase 1 clinical trial in which LT recipients diagnosed with definite or probable AMR according to the 2016 ISHLT definition (23) (n = 18) will be treated with escalating doses of Siltuximab plus standard of care (SOC) immunosuppressive treatment for AMR and baseline maintenance immunosuppression. We will use a traditional 3 + 3 Phase 1 clinical trial design (Figure 2). This safe and easy to implement design proceeds with cohorts of 3 participants:
Cohort A
In Cohort A, 3 participants will be treated with Siltuximab 5.5 mg/kg for 2 doses on days 1 and 8 (Figure 3). If none of the 3 participants in Cohort A experiences a DLT (Table 3), the next 3 participants will be enrolled in Cohort B (see below). However, if 1 participant in Cohort A experiences a DLT, 3 more participants will be added to Cohort A and treated with the same dose (Figure 2). If no additional participants experience a DLT in Cohort A, the study will progress to Cohort B. If 2 or more of the 6 participants or 2 of the initial 3 participants in Cohort A experience a DLT, the next 3 participants will be enrolled in Cohort A2.
Cohort A2
In Cohort A2, 3 participants will be treated with 5.5 mg/kg for 2 doses on days 1 and 22. If none of the 3 participants in Cohort A2 experiences a DLT, the study will progress to Cohort B. If 1 participant in Cohort A2 experiences a DLT, 3 more participants will be added to Cohort A2 and treated with the same dose.
Cohort B
In Cohort B, 3 participants will be treated with Siltuximab 11 mg/kg for 2 doses on days 1 and 22 (Figure 3). If none of the 3 participants in Cohort B experiences a DLT, the next 3 participants will be enrolled in Cohort C. If 1 participant in Cohort B experiences a DLT, 3 more participants will be added to Cohort B and treated with the same dose. If no additional participants experience a DLT in Cohort B, the study will progress to Cohort C. If 2 or more of the 6 participants or 2 of the initial 3 participants in Cohort B experience a DLT, the maximum tolerated dose (MTD) will be designated as that used in Cohort A (or Cohort A2 if that was the previous Cohort before Cohort B).
Cohort C In Cohort C, 3 subjects will be treated with Siltuximab 11 mg/kg for 2 doses on days 1 and 8 (Figure 3). If none of the 3 participants in Cohort C experiences a DLT, the next 3 participants will be enrolled in Cohort D. If 1 participant in Cohort C experiences a DLT, 3 more participants will be added to Cohort C and treated with the same dose. If no additional participants experience a DLT in Cohort C, the study will progress to Cohort D. If 2 or more of the 6 participants or 2 of the initial 3 participants in Cohort C experience a DLT, the MTD will be designated as that used in Cohort B.
Cohort D
In Cohort D, 3 subjects will be treated with Siltuximab 11 mg/kg on days 1, 8, and 22 for a total of 3 doses (Figure 3). If none of the 3 participants in Cohort D experiences a DLT, we will designate this as the MTD. If 1 participant in Cohort D experiences a DLT, 3 more participants will be added to Cohort D and treated with the same dose. If no additional participants experience a DLT, we will consider this the MTD. If 2 or more of the 6 participants or 2 of the 3 initial participants in Cohort D experience a DLT, the MTD will be designated as that used in Cohort C.
The MTD
The MTD is the highest dosing schedule where < 33% of patients experience a DLT. Once the MTD is identified, we will enrol additional patients to reach a total number of 18 (in all cohorts) and treat them with this dosing schedule to confirm MTD and develop preliminary data to inform the design of a future Phase 2 clinical trial. The approved dose of Siltuximab for MCD is 11 mg/kg intravenously every 3 weeks. However, the optimal dose and dosing schedule for the treatment of AMR after LT are not known. The role of IL-6 and its concentrations in serum and local tissues is different in different disease states. Indeed, IL-6 concentrations in serum are generally higher in MCD than in AMR. In one study, patients with MCD had median serum IL-6 concentrations of 24.5 (range: 6.5-93) pg/mL whereas patients with AMR have median serum IL-6 concentrations of 2.20 (range: 1.58-5.06) pg/mL (78). Likewise, IL-6 concentrations in hyperplastic lymph nodes in MCD are likely different than in the lung allograft in AMR. Therefore, it is possible that lower doses of Siltuximab are needed for the treatment of AMR. We selected the starting dose of 5.5 mg/kg for 2 doses 1 week apart (Cohort A) although this dose did not suppress CRP to undetectable levels in MCD because it is possible that it may be sufficient in AMR (62, 74). In Cohort B, we will examine the safety and pharmacodynamics of the dose approved in MCD (11 mg/kg every 3 weeks for 2 doses). In Cohort C, we will examine an expedited dosing schedule (11 mg/kg on days 1 and 8 for 2 doses) to assess if this more rapid suppression of IL-6 activity is safe. Finally, we will assess the safety of 3 doses in Cohort D. 4.2. Primary endpoint
The primary endpoint is safety and tolerability which will be assessed based on the development of dose limiting toxicities (DLT), adverse events (AE), and serious adverse events (SAE). Table 3 below details and defines the different DLT.
Table 3 - Dose limiting toxicities
The development of DLT will be assessed during the DLT follow-up period which is defined as 30 days after the last dose of Siltuximab (Figure 3). All DLT, AE, and SAE will be reviewed by the CEAC to adjudicate if a DLT has occurred. Where applicable, the study will use the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0. The study defines a hypersensitivity reaction as a DLT if this meets CTCAE Grade > 3 allergic reaction. An allergic reaction is defined as a disorder characterized by an adverse local or general response from exposure to an allergen. Grade 3 allergic reaction is one that results in bronchospasm, requires hospitalization, or intravenous intervention. Death is defined as a DLT if the CEAC adjudicates that death was due to infection because approximately 20% of LT recipients with AMR die within 30 days of the diagnosis without treatment with IL-6 signaling blockade (20). Grade 3 thrombocytopenia (platelet count < 50,000 cells/mm3) and Grade 4 neutropenia (absolute neutrophil count < 500 cells/mm3) lasting at least 3 days are defined as DLT. Grade 3 rash is defined as a DLT if it covers > 30% of the body surface area (BSA) with moderate or severe symptoms limiting self-care. Grade 3 pruritis is defined as a DLT if it is widespread and constant, limits self-care, or requires systemic steroids. Hyperlipidemia is defined as a DLT if it results in pancreatitis or requires hospitalization or prolongs a hospitalization. Infections are defined as DLT if they require hospitalization or prolong a hospitalization. Typically, requirement for intravenous antibiotics is the threshold for Grade 3 infections in the CTCAE. However, LT recipients are frequently treated with intravenous antibiotics for non-serious upper or lower respiratory tract infections with antibiotic-resistant organisms. Yet, the use of intravenous antibiotics in this setting is not an indication of the clinical severity of infection. Thus, we have modified the definition of infection as a DLT to be more applicable to LT recipients.
4.3. Secondary Endpoints
See section 1.4 above.
4.4. Exploratory Endpoints
See section 1.5 above.
5. Selection of Participants and Sites
5.1. Lung transplant recipients with AMR
The study will enrol adult lung transplant recipients who develop probable or definite AMR according to the 2016 ISHLT definition (23). The study will not enrol children as the lung transplant programs enrolling participants do not provide care for children. Lung transplant recipients who develop AMR have a poor prognosis. IL-6 plays an important role in mediating allograft injury, and IL-6 blockade might mitigate this. The eligibility criteria for enrolment have been determined based on known adverse events associated with IL-6 blockade and Siltuximab, the package insert, and the risk of infection associated with augmented immunosuppression.
For inclusion and exclusion criteria, see sections 1.6 and 1.7 above.
6. Investigational Agent
6.1. Siltuximab (Sylvant®)
Siltuximab is the investigational agent in this study. Details describing the study related product, labeling, supply, storage, preparation, dispensing, monitoring and accountability are described below. Siltuximab is manufactured by EUSA Pharma and is approved by the FDA as detailed below. Siltuximab is a first-in-class chimeric (humanmouse) immunoglobulin G1K (IgGlK) monoclonal antibody against human IL-6 in a Chinese hamster ovary cell line.
6.2. Dosage and administration
The Siltuximab dose is given as an intravenous infusion over 60 minutes. This Phase 1 study will assess the safety and tolerability of 4 dosing schedules to determine the MTD (see Section 4.1 above and Figure 3). Complete blood count should be checked before each dose of Siltuximab. Before the first administration, the absolute neutrophil count must be > 1,000 cells/mm3, the platelet count must be > 75,000 cells/mm3, and the hemoglobin must be < 17 g/dL. Before subsequent doses, the absolute neutrophil count must be > 1,000 cells/mm3, the platelet count must be > 50,000 cells/mm3, and the hemoglobin must be < 17 g/dL. If these criteria are not me, the dose may be delayed but the dose should not be reduced. Participants will be premedicated 30-60 minutes prior to the start of the Siltuximab infusion using:
1. Acetaminophen 650 or 1000 mg by mouth,
2. Diphenhydramine 25 or 50 mg by mouth, and
3. Methylprednisolone 60 mg intravenously.
6.3. Instructions to followed in the study for preparation and administration Use aseptic technique for reconstitution and preparation of dosing solution. Parenteral drug products should be inspected visually for particulate matter and discoloration prior to administration, whenever solution and container permit.
1. Calculate the dose (mg), total volume (mL) of reconstituted Siltuximab solution required and the number of vials needed. A 21-gauge 1.5-inch needle is recommended for preparation. Infusion bags (250 mL) must contain Dextrose 5% in water and must be made of polyvinyl chloride (PVC), polyolefin (PO), polypropylene (PP), or polyethylene (PE).
2. Allow the vial(s) of Siltuximab to come to room temperature over approximately 30 minutes. Siltuximab should remain at room temperature for the duration of the preparation.
3. Aseptically reconstitute each Siltuximab vial as below: powder. Do not shake or swirl vigorously. Do not remove the contents until all the solids have been completely dissolved. The lyophilized powder should dissolve in less than 60 minutes. Once reconstituted, and prior to further dilution, inspect the vials for particulates and discoloration. Do not use if particles or solution discoloration are present or if visibly opaque. The reconstituted product should be kept for no more than 2 hours prior to addition into the infusion bag.
4. Dilute the reconstituted Siltuximab solution dose to 250 mL with sterile Dextrose 5% in water by withdrawing a volume equal to the total calculated volume of reconstituted Siltuximab from the Dextrose 5% in water, 250 mL bag. Slowly add the total calculated volume (mL) of reconstituted Siltuximab solution to the Dextrose 5% in water infusion bag. Gently invert the bag to mix the solution.
5. Administer the diluted Siltuximab solution in 5% Dextrose in water 250 mL by intravenous infusion over a period of 1 hour using administration sets lined with PVC, or polyurethane (PU), or PE, containing a 0.2-micron inline polyethersulfone (PES) filter. The infusion should be completed within 4 hours of the dilution of the reconstituted solution to the infusion bag.
6. Do not infuse Siltuximab concomitantly in the same intravenous line with other agents.
7. Siltuximab does not contain preservatives. Do not store any unused portion of the reconstituted product or of the infusion solution. Waste material should be disposed of in accordance with local requirements.
6.4. Infusion supervision
Siltuximab will be administered in a hospital or outpatient infusion center where full resuscitation facilities are immediately available and under close supervision of the investigator or designated safety accessor. The intravenous infusion will be supervised by the clinical staff (e.g., clinical nurse or physician) at the participating institutions. A history of each infusion and any adverse side effects will be recorded and reported using appropriated case report forms (CRF). Prior to each infusion, clinical labs (ANC, platelets, bilirubin, ALT, and AST) will be completed. Results must be available prior to initiating the infusion. Vital signs (temperature, blood pressure, pulse, and respiratory rate) will be obtained prior to the start of each infusion and every 15 minutes until the end of the infusion, and every hour thereafter for a total of 2 hours after the first infusion, and 1 hour after subsequent infusions. After the infusion, the intravenous line should remain in the participant for at least 1 hour to enable the administration of drugs if necessary. Additional vital signs may be obtained as clinically indicated. 6.5. Instructions regarding siltuximab dosing delays and discontinuation criteria
6.5.1. Serious infections
Closely monitor patients for the development of signs and symptoms of infection during and after treatment with Siltuximab, as signs and symptoms of acute inflammation may be lessened due to suppression of the acute phase reactants. Hold Siltuximab if a patient develops a serious infection, an opportunistic infection, or sepsis. A patient who develops a new infection during treatment with Siltuximab should undergo a prompt and complete diagnostic workup appropriate for an immunocompromised patient, initiate appropriate antimicrobial therapy, and closely monitor the patient. Siltuximab may be resumed at the next scheduled dose timepoint if the infection has resolved.
6.5.2. Neutropenia
Treatment with Siltuximab was associated with a higher incidence of neutropenia. Infections have been uncommonly reported in association with treatment-related neutropenia in studies and post marketing clinical experience. Per package insert, it is not recommended to initiate Siltuximab in patients with an ANC < 1000 cells/mm3. If the ANC is < 1000 cells/mm3, treatment with Siltuximab may be delayed, but the dose should not be reduced. For this study, if the ANC is < 1000 cells/mm3, the scheduled dose of Siltuximab may be delayed up to 3 days. If during this time the ANC is > 1000 cells/mm3, the dose may be given. However, if the ANC remains < 1000 cells/mm3, the dose will not be administered.
6.5.3. Thrombocytopenia
Treatment with Siltuximab was associated with a reduction in platelet count. Treatment-related reduction in platelets was not associated with serious bleeding events in clinical trials. Per the package insert, it is not recommended to initiate Siltuximab in patients with a platelet count < 75,000 cells/mm3, and it is not recommended to receive subsequent doses of Siltuximab in patients with a platelet count < 50,000 cells/mm3. For this study, if the platelet count is < 50,000 cells/mm3, the scheduled dose of Siltuximab may be delayed up to 3 days. If during this time the platelet count is > 50,000 cells/mm3, the dose may be given. However, if the platelet count remains < 50,000 cells/mm3, the dose will not be administered.
6.5.4. Polycythemia
Per the package insert, Siltuximab may increase hemoglobin levels in patients with MCD. Treatment with Siltuximab is not recommended in patients with a hemoglobin > 17 g/dL. For this study, if the hemoglobin is > 17 g/dL, the scheduled dose of Siltuximab may be delayed up to 3 days. If during this time the hemoglobin is < 17 g/dL, the dose may be given. However, if the hemoglobin remains > 17 g/dL, the dose will not be administered.
6.5.5. Siltuximab discontinuation
Siltuximab will be discontinued in the following circumstances:
1. Participant's decision,
2. Anaphylaxis or other hypersensitivity reaction,
3. GI tract perforation,
4. Pregnancy during the study treatment period,
5. The site PI believes that study therapy is no longer in the best interest of the participant (e.g., due to medical decision, pneumonia, serious infection, opportunistic infection).
In the event that treatment with Siltuximab is discontinued, participants will be followed for the duration of the study period (90 days after enrollment), and data from all safety assessments will be collected.
7. Other Medications
7.1. Maintenance immunosuppression
In general, lung transplant recipients are treated with a combination of Tacrolimus, Mycophenolate Mofetil (MMF), and Prednisone for maintenance immunosuppression. In some cases, Cyclosporine A is substituted for Tacrolimus or Azathioprine is substituted for MMF because of toxicity. During this study, participants will continue their maintenance immunosuppressive regimen, but Prednisone will be held as they receive intravenous Methylprednisolone.
7.2. Standard of care treatment for AMR
Lung transplant recipients with AMR are routinely hospitalized because of allograft dysfunction and to initiate intensive immunosuppressive treatment. Depending on the clinical course and response to treatment, some patients may be discharged from the hospital to complete treatment in the outpatient infusion center. Standard of care (SOC) treatment for AMR varies based on patient-specific factors including overall performance status, previous rejection treatments, lung biopsy findings at the time of AMR diagnosis, and history of infection. The most common treatment combination consists of Carfilzomib, rabbit anti-thymocyte globulin (ATG), high-dose corticosteroids, and IVIG. Carfilzomib 20 mg/m2 is given on days 1, 2, 8, 9, 15, and 16, ATG (1-1.5 mg/kg) is given on days 1-5 targeting a cumulative dose of 5-7.5 mg/kg, methylprednisolone 1 mg/kg is given daily for 7-10 days with a tapering corticosteroid schedule based on clinical response, and IVIG 500-1000 mg/kg is first given on day 10 and continued monthly thereafter. PLEX is not routinely used in the management of AMR because of lack of efficacy and confounding the dosing of antibody treatments including ATG and IVIG. In some cases, a single dose of Rituximab 375 mg/m2 may be administered instead of ATG or for refractory disease at least 72 hours apart from the first dose of IVIG. After enrolling in the study, subjects will receive the first dose of Siltuximab intravenously on day 1. Siltuximab will be given on the same day as the first dose of Carfilzomib and rabbit ATG. These would all be administered in the hospital. Subsequent doses may be given either during the hospitalization or in an outpatient infusion center depending on the patient's clinical status.
7.3. Prophylactic medications
Participants at risk for CMV (i.e., seronegative recipients of seropositive donors or seropositive recipients) will be treated with Valganciclovir prophylactically after treatment for AMR. Similarly, all lung transplant recipients are maintained on prophylaxis for Pneumocystis jirovecii with either Trimethoprim-Sulfamethoxazole or an alternative drug if allergic to sulfa. All patients being evaluated for AMR undergo bronchoscopy with lung biopsies, bronchial washings, and/or bronchoalveolar lavage as part of the routine clinical evaluation. Bronchoscopy specimens are routinely tested for mycobacterial, fungal, and bacterial organisms as well as polymerase chain reaction (PCR) testing for CARV. Although positive mycobacterial, fungal, or bacterial cultures do not necessarily represent invasive disease or infection, patients are routinely started on appropriate antimicrobial therapy before initiating treatment for AMR to minimize the risk of invasive disease and infection after treatment for AMR. Patients who have positive cultures for a mold from bronchoscopy specimens are routinely treated with Voriconazole, Posaconazole, or Isavuconazonium. Likewise, patients who have a positive bacterial culture are treated with appropriate antimicrobials. For patients who have a positive mycobacterial culture, anti-mycobacterial therapy is initiated depending on the specific organism. However, patients who have a positive mycobacterial culture with a potentially pathogenic organism (e.g., Mycobacterium abscessus, Mycobacterium avium complex) will not be eligible for enrolment.
7.4. Rescue treatments
Participants may require rescue treatments for progressive or refractory allograft dysfunction due to AMR. Rescue treatments for AMR may include plasma exchange and Eculizumab, a monoclonal antibody to complement component (C5). The decision to add one of these treatments will be made by the treating lung transplant clinician. If during the course of therapy with Siltuximab a participant requires rescue treatment, subsequent doses of Siltuximab will not be given, and the patient will continue study follow-up per protocol. The treating lung transplant clinician may decide to use rescue therapy with Tocilizumab for progressive or refractory AMR. There are limited data in the literature about the use of rescue Tocilizumab after treatment with Siltuximab. The data are in the setting of cytokine release syndrome (CRS) or immune effector cell associated neurotoxicity syndrome (ICANS) after chimeric antigen receptor (CAR) T-cell therapy (79). In this setting, rescue Tocilizumab has been given for persistent grade 1 CRS at 7 days after the first dose of Siltuximab or at 72 hours for grade 2 CRS. In this study, rescue Tocilizumab may be given 4 half-lives (64 days) after the last administered dose of Siltuximab.
8. Study Procedures
8.1. Informed consent
Sites will identify potential study participants from their cohorts of lung transplant recipients. The research study will be explained in lay terms to each potential participant, and the potential participant will sign an informed consent form before undergoing any study procedures. The investigator or study research coordinator will meet with the study candidate to review all the required elements of informed consent. After the informed consent has been signed, and the participant meets eligibility criteria, the participant will be enrolled in the study.
8.2. Study follow-up and testing
All study participants will be followed for 90 days after enrolment. Participants will be followed more closely for 30 days after the last dose of Siltuximab to assess for DLT. Participants in Cohorts A and C receive the last dose of Siltuximab on day 8 and will be followed for DLT through day 38. Participants in Cohorts B and D receive the last dose of Siltuximab on day 22 and will be followed for DLT through day 52. If Cohort A2 is necessary, participants in this cohort receive the last dose of Siltuximab on day 22 and will be followed for DLT through day 52. The schedule of events for each cohort is shown in Appendix 1.
During the hospitalization, participants will be assessed on a daily basis. This includes a comprehensive History and Physical Exam and review of daily lab work results to assess for treatment-related AE and DLT. On days where testing and Siltuximab dosing are both scheduled, testing will be performed first. Accurate measurements of serum IL-6 concentrations by ELISA during treatment with Siltuximab is not feasible. Thus, CRP will be a surrogate measurement of IL-6 activity (76, 77). IL-6 bioavailability will be assessed by reporter gene assay (RGA) which employs an IL-6-responsive reporter cell line (DS-l/SIE-luciferase) that has been shown to have high dynamic range and specificity for IL-6 blockade in the setting of treatment with anti-IL-6 or anti-IL-6R monoclonal antibodies (80). Additional study-related testing includes baseline serum and exhaled breath condensate (EBC) IL-6 concentrations, and serial measurements of donor-derived cfDNA, DSA, and spirometry as functional biologic assessments of treatment outcome. EBC IL-6 concentrations will be assessed as an exploratory metric at baseline to assess intra-pulmonary levels as serum levels may not reflect intrapulmonary production (57, 58, 60). EBC IL-6 concentrations are significantly higher among cigarette smokers and patients with chronic obstructive pulmonary disease (COPD) than healthy controls but have not been measured in LT recipients (81, 82). Fasting lipids and uric acid levels may be affected by IL-6 blockade and will be measured serially.
9. Criteria for Participant Completion, Withdrawal, and Replacement
9.1. Participant completion
Participants have completed the study when they have completed the final visit on day 90.
9.2. Participant withdrawal
Participants may be withdrawn from the study for the following reasons:
1. The participant elects to withdraw consent from all future study activities including follow-up.
2. The participant is lost to follow-up (i.e., no further follow-up is possible because attempts to reestablish contact with the participant have failed).
3. The participant dies.
9.3. Participant replacement
In the event that a subject is withdrawn for a reason other than a DLT or death (of any cause), another subject will be enrolled into that cohort. Advancement to a successive dosing cohort will not proceed until at least 3 subjects have completed 30 days of follow-up after the last dose of Siltuximab.
10. Safety Monitoring and Reporting
10.1. Safety Definitions
10.1.1. Adverse event (AE)
Any untoward or unfavorable medical occurrence associated with the subject's participation in the research, whether or not considered related to the subject's participation in the research (modified from the definition of adverse events in the 1996 International Conference on Harmonization E-6 Guidelines for Good Clinical Practice) (from CHRP "Guidance on Reviewing and Reporting Unanticipated Problems Involving Risks to Subjects or Others and Adverse Events" http://www.hhs.gOv/ohrp/policy/advevntguid.html#Q2 ).
10.1.2. Suspected adverse reaction (SAR)
An SAR is any adverse event for which there is a reasonable possibility that the study drug caused the adverse event. For the purposes of safety reporting, "reasonable possibility" means there is evidence to suggest a causal relationship between the drug and the adverse event. An SAR implies a lesser degree of certainty about causality than adverse reaction, which means any adverse event caused by a drug.
10.1.3. Unexpected adverse event
An adverse event or suspected adverse reaction is considered "unexpected" if it is not listed in the most recent version of the Siltuximab Investigator Brochure (IB) or is not listed at the specificity, severity or rate of occurrence that has been observed as described in the IB; or is not consistent with the risk information described in the general investigational plan or elsewhere in the IND. "Unexpected" also refers to adverse events or suspected adverse reactions that are mentioned in the IB as occurring with a class of drugs or as anticipated from the pharmacological properties of the drug but are not specifically mentioned in the IB as occurring with the particular drug under investigation.
10.1.4. Serious adverse event
An adverse event or suspected adverse reaction is considered "serious if, in the view of the investigator, it results in any of the following outcomes:
1. Death,
2. A life-threatening event: an AE or SAR is considered "life-threatening" if, in the view of the investigator, its occurrence places the subject at immediate risk of death. It does not include an AE or SAR that, had it occurred in a more severe form, might have caused death,
3. Inpatient hospitalization or prolongation of existing hospitalization,
4. Persistent or significant incapacity or substantial disruption of the ability to conduct normal life functions,
5. Congenital anomaly or birth defect,
6. Important medical events that may not result in death, be life threatening, or require hospitalization may be considered serious when, based upon appropriate medical judgment, they may jeopardize the subject and may require medical or surgical intervention to prevent one of the outcomes listed above.
Elective hospitalizations or hospital admissions for the purpose of conduct of protocol mandated procedures are not to be reported as an SAE unless hospitalization is prolonged due to complications.
10.2. Grading of adverse events
The study site will grade the severity of adverse events experienced by participants according to the criteria set forth in NCI-CTCAE, version 5.0. This document provides a common language to describe levels of severity, to analyze and interpret data, and to articulate the clinical significance of all adverse events. Adverse events will be graded on a scale from 1 to 5 according to the following standards in the NCI-CTCAE manual:
Table 4 - Grading of adverse events
Events grade > 3 will be recorded on the appropriate AE case report form for this study. For grading an abnormal value or result of a clinical or laboratory evaluation, a treatment-emergent adverse event is defined as an increase in grade from baseline or from the last post-baseline value that doesn’ t meet grading criteria. Changes in grade from screening to baseline will also be recorded as adverse events but are not treatment emergent. If a specific event or result from a given clinical or laboratory evaluation is not included in the NCI-CTCAE manual, then an abnormal result would be considered an adverse event if changes in therapy or monitoring are implemented because of the event/result. Example 3: A Phase 1 Clinical Trial of Siltuximab for the Treatment of Antibody-Mediated Rejection after Lung Transplantation (Updated)
The following Phase 1 Clinical Trial protocol is based on the protocol set out in Example 2 but has been updated after consultation with regulatory agencies.
This is a Phase 1, two-center, double-blind, randomized placebo-controlled, clinical trial in which 30 lung transplant recipients with antibody-mediated rejection will be randomized 1: 1 : 1 to low dose Siltuximab, full dose Siltuximab, or Placebo intravenously on days 1 and 22 in addition to routine clinical treatment for antibody- mediated rejection.
Study Arm 1 : low dose Siltuximab (n = 10)
• Siltuximab 5.5 mg/kg intravenously on days 1 and 22 Study Arm 2: full dose Siltuximab (n = 10)
• Siltuximab 11 mg/kg intravenously on days 1 and 22 Study Arm 3: Placebo (n = 10)
• Placebo intravenously on days 1 and 22
1. Protocol synopsis
1.1. Primary Objectives
See Example 2, Section 1.1.
1.2. Secondary Objectives
The secondary objectives are:
1. Assess the pharmacokinetics of Siltuximab after dose 1 and dose 2 of treatment,
2. Assess the pharmacodynamics of Siltuximab after dose 1 and dose 2 of treatment,
3. Determine the optimal dose for future trials,
4. Assess functional biological measurements and clinical outcomes that may be used to assess efficacy in future trials.
1.3. Primary Endpoint
The primary endpoint is the incidence of CTCAE > grade 3 during a period of 90 days after randomization.
1.4. Secondary Endpoints
The following secondary endpoints will be assessed: 1. The incidence of CTCAE > grade 3 during a period of 180 days after randomization,
2. Undetectable serum high-sensitivity C-reactive protein (HS-CRP) (< 1 mg/L) 90 days after randomization,
3. Undetectable serum high-sensitivity C-reactive protein (HS-CRP) (< 1 mg/L) 180 days after randomization,
4. Siltuximab concentrations between randomization and day 90,
5. Clearance of donor-specific antibodies (DSA) between randomization and day 180,
6. Hyperuricemia (> 7 mg/dL) between randomization and day 180,
7. Hyperlipidemia requiring the initiation or dose increase of medical therapy between randomization and day 180,
8. Confirmed bacterial, cytomegalovirus (CMV) (defined as a positive blood viral load > 200 lU/mL), mold, mycobacterial, or community-acquired respiratory viral infection (each assessed independently) between randomization and day 180,
9. Opportunistic infection between randomization and day 180,
10. Change in Forced Vital Capacity (FVC) between randomization and day 180,
11. Change in Forced Expiratory Volume in One Second (FEV1) between randomization and day 180,
12. Development of probable chronic lung allograft dysfunction (CLAD) according to the 2019 ISHLT criteria between randomization and day 180,
13. Allograft survival defined as death or undergoing re-transplantation between randomization and day 180.
1.5. Exploratory Endpoints
The following exploratory endpoints will be assessed:
1. Donor-derived cell-free-DNA < 1%,
2. Change in circulating donor-derived cell-free-DNA after study drug treatment,
3. Change in HS-CRP between randomization and day 180,
4. Serum IL-6 levels at enrollment only (before administration of the first dose of Siltuximab),
5. Exhaled breath condensate IL-6 levels at enrollment,
6. Serial measurements of IL-6 bioavailability by reporter gene assay (RGA).
1.6. Inclusion criteria
Individuals who meet all the following criteria are eligible for enrolment as study participants: 1. 18 years of age or older,
2. Single or bilateral lung transplant recipient,
3. New diagnosis of clinical definite, probable, or possible antibody-mediated rejection according to the 2016 International Society for Heart and Lung Transplantation (ISHLT) definition with plans to be treated with Carfilzomib and/or anti-thymocyte globulin,
4. Admitted to the hospital for treatment of AMR,
5. Donor-specific antibodies (DSA) to human leukocyte antigens (HLA) with a Mean Fluorescence Intensity (MFI) > 1000,
6. Able to understand the purpose of the study and willing to participate and sign informed consent.
1.7. Exclusion Criteria
Individuals who meet any of these criteria are not eligible for enrolment as study participants.
1. Pregnant or breast feeding,
2. Airway anastomotic dehiscence on bronchoscopy,
3. Thoracotomy incision dehiscence,
4. Underwent lung transplantation less than 6 months before enrollment,
5. Underwent other invasive surgical procedure less than 6 weeks before enrollment,
6. History of lymphoma or hematologic malignancy,
7. Treatment with IL-6 signaling blockade with 6 months of enrollment,
8. Planned treatment with plasma exchange (PLEX) for AMR,
9. Cancer other than non-melanoma skin cancer with disease-free period <3 years,
10. Positive respiratory virus PCR detected within 7 days of enrollment,
11. Active cytomegalovirus infection within 7 days of enrollment,
12. Positive respiratory culture for Mycobacterium tuberculosis, Mycobacterium abscessus, Mycobacterium chelonae, or Mycobacterium avium complex within 4 weeks of enrollment,
13. Absolute neutrophil count (ANC) < 1,000 cells/mm3 at enrollment,
14. Platelet count < 75,000 cells/mm3 at enrollment,
15. Hemoglobin > 17 g/dL at enrollment,
16. ALT or AST > 2.5 times upper limit of normal at enrollment,
17. Total bilirubin > 2.5 times upper limit of normal at enrollment,
18. Uric acid > 7 mg/dL at enrollment.
19. History of gastrointestinal tract perforation,
20. History of diverticulitis (diverticulosis is not an exclusion), 21. Plan for surgical procedure (other than bronchoscopy) within 120 days of enrollment.
22. Inability or unwillingness to give written informed consent or comply with the study protocol,
23. Any condition that in the opinion of the site investigator introduces undue risk by participating in this study or impacts the quality or interpretation of the study results.
1.8. Study Stopping Rules
See Example 2, Section 1.8. Selected adverse events of concern and their thresholds in this trial are:
1. Any occurrence of gastrointestinal tract perforation,
2. Death in 20% or more of enrolled subjects,
3. Opportunistic infections in 25% or more of enrolled subjects.
2. Study Definitions
See the definitions in Example 2, Section 2, in addition to those below.
Serious infection: Infection requiring intravenous antimicrobial therapy and hospitalization.
Tissue invasive CMV: Tissue invasive CMV is defined as the presence of signs and symptoms of end-organ disease such as enteritis, colitis, pneumonitis, retinitis and CMV replication in blood.
In this study, Antibody-mediated rejection will be defined based on the 2016 ISHLT definition. Only cases of clinical definite, probable, or possible AMR with donor-specific antibodies (DSA) to human leukocyte antigens (HLA) will be considered as AMR in the study.
Moreover, in this study, further to the definition in Example 2, Section 2, the definition of opportunistic infection may include pathogenic non-tuberculous mycobacteria (NTM).
3. Study Hypothesis
We hypothesize that the addition of Siltuximab to routine clinical treatment for AMR is sufficiently safe and clinically tolerable to proceed with a subsequent Phase 2 clinical trial that assesses efficacy. To test this hypothesis, we will conduct a Phase 1 clinical trial of 30 adult lung transplant recipients with AMR. 4. Study Design
4.1. Description of study design
This is a Phase 1, two-center, double-blind, randomized, placebo-controlled clinical trial in which 30 LT recipients diagnosed with definite or probable AMR according to the 2016 ISHLT definition (23) will be randomized in a 1: 1 : 1 ratio to receive low dose Siltuximab, full dose Siltuximab, or Placebo (Dextrose 5% in sterile water) intravenously on days 1 and 22 in addition to routine clinical treatment for AMR and baseline maintenance immunosuppression. The placebo arm enhances the detection of safety signals by serving as a control group to compare AE rates in the Siltuximab group to.
Study Arm 1 : low dose Siltuximab (n = 10)
• Siltuximab 5.5 mg/kg intravenously on days 1 and 22 (two total doses)
• Routine clinical treatment for AMR
• Baseline maintenance immunosuppression
Study Arm 2: full dose Siltuximab (n = 10)
• Siltuximab 11 mg/kg intravenously on days 1 and 22 (two total doses)
• Routine clinical treatment for AMR
• Baseline maintenance immunosuppression Study Arm 3: Placebo (n = 10)
• Placebo (sterile D5W) intravenously on days 1 and 22 (two total doses)
• Routine clinical treatment for AMR
• Baseline maintenance immunosuppression
The study will use safety, pharmacokinetic, pharmacodynamic, functional biological measurements, and clinical outcomes data to determine an optimal dose for a future Phase 2 clinical trial that assesses the efficacy of Siltuximab in the treatment of AMR after LT. Although, the FDA-approved dose of Siltuximab for MCD is 11 mg/kg intravenously every 3 weeks, the optimal dose for the treatment of AMR after LT is not known. The role of IL-6 and its concentrations in serum and local tissues is different in different disease states. Indeed, IL-6 concentrations in serum are generally higher in MCD than in AMR. In one study, patients with MCD had median serum IL-6 concentrations of 24.5 (range: 6.5-93) pg/mL whereas patients with AMR have median serum IL-6 concentrations of 2.20 (range: 1.58-5.06) pg/mL (78). Likewise, IL-6 concentrations in hyperplastic lymph nodes in MCD are likely different than in the lung allograft in AMR. Therefore, it is possible that lower doses of Siltuximab are effective for the treatment of AMR. We selected the low dose of 5.5 mg/kg for 2 doses 21 days apart although this dose did not suppress CRP to undetectable levels in MCD because it is possible that it may be sufficient in AMR (62, 74). We will also examine the safety, pharmacokinetics, and pharmacodynamics of the dose approved in MCD (11 mg/kg every 21 days for 2 doses).
4.2. Primary Endpoint
See Section 1.3 above.
4.3. Secondary Endpoints
See Section 1.4 above.
4.4. Exploratory Endpoints
See Section 1.5 above.
4.5. Randomization and blinding
Eligible subjects will be randomized on the day of enrollment through a web-based randomization system with a 1: 1 : 1 ratio to low dose Siltuximab, full dose Siltuximab, or Placebo. The randomization list will be computer generated using a permuted block randomization and stratified by enrollment site. Treatment allocation will remain concealed except to each site's study pharmacist and unblinded statistician. The study staff at each site, the participants, and the clinical team will remain blinded to treatment assignment.
5. Selection of Participants and Sites
5.1. Lung transplant recipients with AMR
The study will enroll adult lung transplant recipients who develop clinical definite, probable, or possible AMR according to the 2016 ISHLT definition (23) and DSA to HLA. The study will not enroll children as the lung transplant programs enrolling participants do not provide care for children. Lung transplant recipients who develop AMR have a poor prognosis. IL-6 plays an important role in mediating allograft injury, and IL-6 blockade might mitigate this. The eligibility criteria for enrollment and randomization have been determined based on known adverse events associated with IL-6 blockade and Siltuximab, the package insert, and the risk of infection associated with augmented immunosuppression.
See Sections 1.6 and 1.7 for inclusion and exclusion criteria respectively.
6. Investigational Agent The investigational agents for this trial are Siltuximab (Sylvant®) and Placebo (Dextrose 5% in sterile water, D5W). Details describing the study related product labeling, supply, storage, preparation, dispensing, monitoring and accountability are described below.
6.1. Siltuximab (Sylvant®)
See Example 2, Section 6.1. In this study, Siltuximab is manufactured by Recordati Rare Diseases and is approved by the FDA as detailed below.
6.2. Dosage and administration
See Example 2, Section 6.2.
6.3. Instructions for preparation and administration
See Example 2, Section 6.3.
6.4. Placebo
Placebo, D5W, is considered an investigational agent in this study. Sites will use only sterile D5W that is produced by an FDA registered facility as designated by the presence of a National Drug Code number on product labeling. Product information including manufacturer, lot number, and expiration date will be captured in the pharmacy accountability log at each site. Placebo will be prepared with dose volume to match Siltuximab and will be stored using the standard procedures at each site's investigational pharmacy. Placebo should be prepared using appropriate aseptic techniques using the site's D5W stock. Placebo will be prepared and dispensed by an unblinded pharmacist at the site. Placebo will be labeled by the site's investigational pharmacy with the same infusion bag label used for active Siltuximab to maintain the blind.
6.5. Infusion supervision
See Example 2, Section 6.4. The investigational agents in this trial are Siltuximab and a placebo (Dextrose 5% in sterile water, D5W).
6.6. Instructions regarding investigational agent dosing delays and discontinuation
See Example 2, Section 6.5 and subsections. References in this section to "Siltuximab" in relation to actions in this study (i.e. other than references about known facts) should be read as "the investigational agent", in view of the blinding. 7. Other medications
7.1. Maintenance immunosuppression
See Example 2, Section 7.1.
7.2. Routine clinical treatment for AMR and investigational agent administration
Lung transplant recipients with AMR are routinely hospitalized because of allograft dysfunction and to initiate intensive immunosuppressive treatment. Depending on the clinical course and response to treatment, some patients may be discharged from the hospital to complete treatment in the outpatient infusion center. Routine clinical treatment for AMR varies based on patient-specific factors including overall performance status, previous rejection treatments, lung biopsy findings at the time of AMR diagnosis, and history of infection. The most commonly used clinical regimen at the two sites includes high-dose corticosteroids, IVIG, anti-thymocyte globulin (ATG), and Carfilzomib. ATG is not routinely used for induction immunosuppression in the immediate period after lung transplantation at the sites, but it is used to treat persistent ACR and CLAD. Patients who have been previously treated with ATG are eligible for enrollment in this study but will not be treated with an additional course of ATG for AMR; they will be treated with Carfilzomib. All other study participants will be treated with ATG and Carfilzomib. High-dose corticosteroids and IVIG are cornerstone treatments for AMR at the sites, and these will be given to all participants. Thus, study participants will be treated with:
• High-dose corticosteroids,
• IVIG,
• ATG, if it has not been administered previously, and
• Carfilzomib.
Carfilzomib 20 mg/m2 is given on days 1, 2, 8, 9, 15, and 16, ATG (1-1.5 mg/kg) is given on days 1-5 targeting a cumulative dose of 5-7.5 mg/kg, methylprednisolone 1 mg/kg is given daily for 7-10 days with a tapering corticosteroid schedule based on clinical response, and IVIG 500-1000 mg/kg is first given on day 10 and continued monthly thereafter. PLEX is not part of the routine first-line treatment for AMR at the sites because of lack of effectiveness clearing DSA or improving the clinical course. Additionally, PLEX confounds the dosing of antibody treatments including ATG and IVIG and requires the placement of a central intravenous pheresis catheter. Known planned treatment with PLEX at the time of enrollment is an exclusion criterion for this study, but rescue treatment with PLEX for refractory AMR is allowed (see Section 7.4. Rescue treatments below). Subjects will be consented, enrolled, and randomized after admission to the hospital for management of AMR and will receive the first dose of the investigational agent intravenously within 72 hours of admission. Treatment for AMR will not be delayed for study-related reasons. The investigational agent may be given on the same day as the first dose of Carfilzomib and rabbit ATG. These would all be administered in the hospital. The subsequent dose of the investigational agent may be given either during the hospitalization or in an outpatient infusion center depending on the patient's clinical status.
7.3. Prophylactic medications
See Example 2, Section 7.3. In this study, participants at risk for CMB may be prophylactically treated with Letermovir as an alternative to valganciclovir, after treatment for AMR.
7.4. Rescue treatments
See Example 2, Section 7.4. In this study, rescue treatments for AMR may instead include PLEX, Rituximab, and Eculizumab. References in this section to "Siltuximab" should be read as "the investigational agent", in view of the blinding. Moreover, in this study, rescue Tocilizumab may be given after the end of the study (90 days) instead of 64 days after the last administered dose of Siltuximab as in Example 2.
8. Study Procedures
Study procedures including informed consent, History and Physical Exam, study drug dosing, and testing are detailed in the schedule of events for each dosing cohort (Appendix 2).
8.1. Informed consent
See Example 2, Section 8.1. In this study, Patients will be given ample time to decide if they would like to participate.
8.2. Randomization
Randomization will occur after the participant has signed the informed consent form.
8.3. Study follow-up and testing
All study participants will be followed for 180 days after enrollment and randomization, with the primary endpoint occurring at day 90. Participants will be randomized on the same day as enrollment. The investigational agent will be administered on day 1 and day 22. The schedule of events cohort is shown in Appendix 2. During the hospitalization, participants will be assessed on a daily basis. This includes a comprehensive History and Physical Exam and review of daily lab work results to assess for treatment-related AE. On days where testing and investigational agent dosing are both scheduled, testing will be performed first. Accurate measurements of serum IL-6 concentrations by ELISA during treatment with Siltuximab is not feasible. Thus, HS-CRP will be a surrogate measurement of IL-6 activity (76, 77). IL-6 bioavailability will be assessed by reporter gene assay (RGA) which employs an IL-6- responsive reporter cell line (DS-l/SIEIuciferase) that has been shown to have high dynamic range and specificity for IL-6 blockade in the setting of treatment with anti- IL-6 or anti-IL-6R monoclonal antibodies (80). To assess pharmacokinetics, Siltuximab concentrations will be measured using a commercially available ELISA kit specifically designed to measure Siltuximab (Kribiolisa cat # KBI1630) with a minimum detection level of 12 ng/mL and an approximately 100-fold linear detection range. The ELISA kit will also be independently validated with a Siltuximab standard calibration curve generated with healthy human volunteer serum. Timepoints for measuring pharmacokinetics are detailed in the schedule of events above. At every dosing visit, a pre-dose measurement and a post-dose measurement will be made; the post-dose measurement will be made 1 hour after completion of the infusion. To avoid unblinding, pharmacokinetic and HS-CRP measurements will be batched, and the results will only be made available after completion of study follow-up. Additional testing includes baseline serum and exhaled breath condensate (EBC) IL-6 concentrations, and serial measurements of donor-derived cfDNA, DSA, and spirometry as functional biologic assessments of treatment outcome. EBC IL-6 concentrations will be assessed as an exploratory metric at baseline to assess intrapulmonary levels as serum levels may not reflect intrapulmonary production (57, 58, 60). EBC IL-6 concentrations are significantly higher among cigarette smokers and patients with chronic obstructive pulmonary disease (COPD) than healthy controls but have not been measured in LT recipients (81, 82). Fasting lipids and uric acid levels may be affected by IL-6 blockade and will be measured serially. Tests performed for research purposes are HS-CRP, fasting lipids, uric acid, serum and EBC IL-6 concentrations, IL-6 bioavailability, and Siltuximab concentrations. HS-CRP, fasting lipids, and uric acid levels can be measured using the same blood draw as the CMP and do not require additional samples. However, serum IL-6 concentrations, IL-6 bioavailability, and Siltuximab concentrations require separate blood draws; a total of 10 mL of blood will be collected for these research tests at each testing visit. Participants will have 10 mL of blood drawn for research testing at visits 0, 1, 2, 3, 4, 5, 6, 7, and 8 for a total volume of 90 mL during study follow-up. No bronchoscopies or lung biopsies will be performed for study purposes; specifically, the study does not mandate a follow-up biopsy after treatment for several reasons. First, the histologic features of pulmonary AMR are non-specific and improvement in histology has not been defined. Second, patients typically have impaired allograft function after AMR, which increases the risk of serious complications associated with bronchoscopy and biopsy. As a result, a follow-up biopsy after treatment is not done as part of routine clinical care at the sites.
9. Criteria for Participant Completion, Withdrawal, and Replacement
9.1. Participant Completion
Participants have completed the study when they have completed the final visit on day 180.
9.2. Participant withdrawal
Please see Example 2, Section 9.2. In this study participants may be withdrawn from the study if the participant undergoes re-transplantation.
9.3. Participant replacement
After randomization, subjects who withdraw from the study will not be replaced.
10. Safety Monitoring and Reporting
10.1. Safety Definitions
10.1.1. Adverse event, Suspected adverse reaction, Unexpected adverse event and Serious adverse event (SAE)
See Example 2, Section 10.1 and subsections for the definitions of adverse event (AE), suspected adverse reaction (SAR), unexpected adverse event and serious adverse event (SAE).
10.1.2. Serious and unexpected suspected adverse reaction (SUSAR).
A SUSAR is defined as an adverse event that is both serious and unexpected and has a reasonable possibility of a causal relationship with Siltuximab.
10.1.3. Unanticipated problems (UP)
UP are defined as any event that meets all of the following criteria:
1. Unexpected in terms of nature, severity, or frequency given the research procedures and subject population, and
2. Related or probably related to participation in the study or if the event probably or definitely affects the safety, rights and welfare of current participants, and
3. Suggests that the research places subjects at greater risk of harm than was previously recognized. 10.2. Grading of adverse events
See Example 2, Section 10.2. In this study, grading will be performed by the principal investigators (Pls).
ABBREVIATIONS
ACR Acute Cellular Rejection
ADL Activities of Daily Living
AE Adverse Event
ALT Alanine Transaminase
AMR Antibody-Mediated Rejection
ANC Absolute Neutrophil Count
AST Aspartate Aminotransferase
ATG Anti-Thymocyte Globulin
BAL Bronchoalveolar Lavage
BOS Bronchiolitis Obliterans Syndrome
BSA Body Surface Area
BSF-2 B-cell Stimulating Factor 2
C4d Complement Component 4d
CARV Community-Acquired Respiratory Virus
CBC Complete Blood Count
CEAC Clinical Events Adjudication Committee cfDNA Donor-Derived Cell Free-DNA
CLAD Chronic Lung Allograft Dysfunction
CMP Comprehensive Metabolic Profile
CMV Cytomegalovirus
CNI Calcineurin Inhibitor
COPD Chronic Obstructive Pulmonary Disease
CRF Case Report Form
CRP C-Reactive Protein
CRS Cytokine Release Syndrome
CTCAE Common Terminology Criteria for Adverse Events
DCC Data Coordinating Center
DLT Dose Limiting Toxicity
DSA Donor-Specific Antibodies
DSMB Data Safety Monitoring Board
EBC Exhaled Breath Condensate FDA Food and Drug Administration
FEVi Forced Expiratory Volume in One Second
FVC Forced Vital Capacity
GD Gestation Days
GERD Gastro-Esophageal Reflux Disease
GFR Glomerular Filtration Rate
GI Gastrointestinal
GRAfT Genomic Research Alliance for Transplantation gpl30 Glycoprotein 130
HHV-8 Human Herpesvirus-8
HIV Human Immunodeficiency Virus
HLA Human Leukocyte Antigens
HS-CRP High-sensitivity C-reactive protein
IB Investigator Brochure
ICANS Immune Effector Cell Associated Neurotoxicity Syndrome
IDE Investigational Device Exemption
IgGlK Immunoglobulin G1K
IL-6 Interleukin-6
IL-6R Interleukin-6 Receptor
IND Investigational New Drug
IP Investigational Product
IRB Institutional Review Board
ISHLT International Society for Heart and Lung Transplantation
IVIG Intravenous Immune Globulin
JAK Janus Kinase
LT Lung Transplantation
MCD Multicentric Castleman's Disease
MFI Mean Fluorescence Intensity
MM Multiple Myeloma
MMF Mycophenolate Mofetil
MTD Maximum Tolerated Dose
NCI National Cancer Institute
NHP Non-Human Primate
PCR Polymerase Chain Reaction
PGD Primary Graft Dysfunction
PHI Protected Health Information
PI Principal Investigator
PLEX Plasma Exchange PE Polyethylene
PES Polyethersulfone
PO Polyolefin
PP Polypropylene PU Polyurethane
PVC Polyvinyl Chloride
RAS Restrictive Allograft Syndrome
RCT Randomized Controlled Trial
RGA Reporter Gene Assay SAE Serious Adverse Event
SAR Suspected Adverse Reaction
SIL-6R Soluble Interleukin-6 Receptor
SOC Standard of Care
STAT Signal Transducer and Activator of Transcription SUSAR Serious and Unexpected Suspected Adverse Reaction
REFERENCES
1. https://optn.transplant.hrsa.gOv/data/view-data-reports/national-data/#, accessed on August 22, 2022.
2. Chambers DC, Cherikh WS, Harhay MO, et al. The International Thoracic Organ Transplant Registry of the International Society for Heart and Lung Transplantation: Thirty-sixth adult lung and heart-lung transplantation report - 2019; Focus theme: Donor and recipient size match. J Heart Lung Transplant 2019; 38: 1042-1055. PMID 31548030
3. Kulkarni HS, Cherikh WS, Chambers DC, et al. Bronchiolitis obliterans syndrome-free survival after lung transplantation : An International Society for Heart and Lung Transplantation Thoracic Transplant Registry analysis. J Heart Lung Transplant 2019; 38: 5-16. PMC6431291.
4. Verleden GM, Glanville AR, Lease ED, et al. Chronic lung allograft dysfunction : Definition, diagnostic criteria, and approaches to treatment - A consensus report from the Pulmonary Council of the ISHLT. J Heart Lung Transplant 2019; 38: 493-503. PMID 30962148
5. Glanville AR, Verleden GM, Todd JL, et al. Chronic lung allograft dysfunction : Definition and update of restrictive allograft syndrome - A consensus report from the Pulmonary Council of the ISHLT. J Heart Lung Transplant 2019; 38: 483-492. PMID: 31027539
6. Finlen Copeland CA, Snyder LD, Zaas DW, Tubyfill WJ, Davis WA, Palmer SM. Survival after bronchiolitis obliterans syndrome among bilateral lung transplant recipients. Am J Respir Crit Care 2010; 182: 784-9. PMID: 20508211
7. Sato M, Waddell TK, Wagnetz U, et al. Restrictive allograft syndrome (RAS): A novel form of chronic lung allograft dysfunction. J Heart Lung Transplant 2011; 30: 735-42. PMID: 21419659
8. Venado A, Kukreja J, Greenland JR. Chronic lung allograft dysfunction. Thorac Surg Clin 2022; 32: 231-242. PMID: 35512941
9. Gottlieb J, Verleden GM, Perch M, et al. Disease progression in patients with the restrictive and mixed phenotype of chronic lung allograft dysfunction - A retrospective analysis in five European centers to assess the feasibility of a therapeutic trial. PLoS One 2021; 16(12):e0260881. PMID: 34941934
10. Royer P-J, Olivera-Botello G, Koutsokera A, et al. Chronic lung allograft dysfunction: A systematic review of mechanisms. Transplantation 2016; 100: 1803- 1814. PMID: 27257997 11. Bos S, Milross L, Filby AJ, Vos R, Fisher AJ. Immune processes in the pathogenesis of chronic lung allograft dysfunction : Identifying the missing pieces of the puzzle. Eur Respir Rev 2022; 31(165): 220060. PMID: 35896274
12. Cantu E, Appel JZ, Hartwig MG, et al. J. Maxwell Chamberlain Memorial Paper. Early fundoplication prevents chronic allograft dysfunction in patients with gastroesophageal reflux disease. Ann Thorac Surg 2004; 78: 1142-51. PMID 15464462
13. Daud SA, Yusen RD, Meyers BF, et al. Impact of immediate primary lung allograft dysfunction on bronchiolitis obliterans syndrome. Am J Respir Crit Care Med 2007; 175: 507-513. PMID: 17158279
14. Huang HH, Yusen RD, Meyers BF, et al. Late primary graft dysfunction after lung transplantation and bronchiolitis obliterans syndrome. Am J Transplant 2008; 8: 2454- 2462. PMID: 18785961
15. Burton CM, Iversen M, Carlsen J, et al. Acute cellular rejection is a risk factor for bronchiolitis obliterans syndrome independent of post-transplant baseline FEV1. J Heart Lung Transplant 2009; 28: 888-893. PMID: 19716040
16. Glanville AR, Aboyoun CL, Havryk A, et al. Severity of lymphocytic bronchiolitis predicts long-term outcome after lung transplantation. Am J Respir Crit Care Med 2008; 177: 1033-1040. PMID: 18263803
17. Morrell MR, Pilewski JM, Gries CJ, et al. De novo donor-specific HLA antibodies are associated with early and high-grade bronchiolitis obliterans syndrome and death after lung transplantation. J Heart Lung Transplant 2014; 33: 1288-1294. PMID: 25443870
18. Safavi S, Robinson DR, Soresi S, et al. De novo donor HLA-specific antibodies predict development of bronchiolitis obliterans syndrome after lung transplantation. J Heart Lung Transplant 2014; 33: 1273-1281. PMID: 25130554
19. Tikkanen JM, Singer LG, Kim SJ, et al. De novo DQ donor-specific antibodies are associated with chronic lung allograft dysfunction after lung transplantation. Am J Respir Crit Care Med 2016; 194: 596-606. PMID: 26967790
20. Witt CA, Gaut JP, Yusen RD, et al. Acute antibody-mediated rejection after lung transplantation. J Heart Lung Transplant 2013; 32: 1034-1040. PMID: 23953920
21. Aguilar PR, Carpenter D, Ritter J, et al. The role of C4d deposition in the diagnosis of antibody-mediated rejection after lung transplantation. Am J Transplant 2018; 18: 936-944. PMID: 28992372
22. Khalifah AP, Hachem RR, Chakinala MM, et al. Respiratory viral infections are a distinct risk for bronchiolitis obliterans syndrome and death. Am J Respir Crit Care Med 2004; 170: 181- 7. PMID 15130908 23. Levine DJ, Glanville AR, Aboyoun C, et al. Antibody-mediated rejection of the lung: A consensus report of the International Society for Heart and Lung Transplantation. J Heart Lung Transplant 2016; 35: 397-406. PMID 27044531
24. Roux A, Bendib Le Lan I, Holifanjaniaina S, et al. Antibody-mediated rejection in lung transplantation : Clinical outcomes and donor-specific antibody characteristics. Am J Transplant 2016; 16: 1216-1228. PMID: 26845386
25. Otani S, Davis AK, Cantwell L, et al. Evolving experience of treating antibody- mediated rejection following lung transplantation. Transpl Immunol 2014; 31: 75-80. PMID: 25004453
26. Lobo LJ, Aris RM, Schmitz J, Neuringer IP. Donor-specific antibodies are associated with antibody-mediated rejection, acute cellular rejection, bronchiolitis obliterans syndrome, and cystic fibrosis after lung transplantation. J Heart Lung Transplant 2013; 32: 70-77. PMID 23260706
27. Agbor-Enoh S, Jackson AM, Tunc I, et al. Late manifestation of alloantibody- associated injury and clinical pulmonary antibody-mediated rejection: Evidence from cell-free DNA analysis. J Heart Lung Transplant 2018; 37: 925-932. PMID 29500138
28. Jang MK, Tunc I, Berry GJ, et al. Donor -derived cell-free DNA accurately detects acute rejection in lung transplant patients, a multi-center cohort study. J Heart Lung Transplant 2021; 40: 822-830. PMID 34130911
29. Schaper F, Rose-John S. Interleukin-6: Biology, signaling and strategies of blockade. Cytokine Growth Factor Rev 2015; 26: 475-487. PMID: 26189695
30. Rose-John S, Neurath MF. IL-6 trans-signaling: The heat is on. Immunity 2004; 20: 2-4. PMID: 14738759
31. Heink S, Yogev N, Grabers C, et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat Immunol 2017; 18: 74-85. PMID: 27893700
32. Rochman I, Paul WE, Ben-Sasson SZ. IL-6 increases primed cell expansion and survival. J Immunol 2005; 174: 4761-4767. PMID: 15814701
33. Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of pathogenic effector Thl7 and regulatory T cells. Nature 2006; 441 : 235- 238. PMID: 16648838
34. Dong C. Diversification of T-helper-cell lineages: Finding the family root of IL- 17-producing cells. Nat Rev Immunol 2006; 6: 329-333. PMID: 16557264
35. Kishimoto T, Hibi M, Murakami M, Narazaki M, Saito M, Taga T. The molecular biology of interleukin 6 and its receptor. CIBA Found Symp 1992; 167: 5-16. PMID: 1425018 36. Hirano T, Yasukawa K, Harada H, et al. Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin. Nature 1986; 324: 73-76. PMID: 3491322
37. Tanaka T, Kishimoto T. The biology and medical implications of interleukin-6. Cancer Immunol Res 2014; 2: 288-294. PMID: 24764575
38. Ma CS, Deenick EK, Batten M, Tangye SG. The origins, function, and regulation of T follicular helper cells. J Exp Med 2012; 209: 1241-1253. PMID: 22753927
39. Diehl SA, Schmidlin H, Nagasawa M, Blom B, Spits H. IL-6 triggers IL-21 production by human CD4+ T cells to drive STAT3-dependent plasma cell differentiation in B cells. Immunol Cell Biol 2012; 90: 802-811. PMID: 22491065
40. Kawano M, Hirano T, Matsuda T, et al. Autocrine generation and requirement of BSF-2/IL-6 for human multiple myelomas. Nature 1988; 332: 83-85. PMID: 3258060
41. Suematsu S, Matsusaka T, Matsuda T, et al. Generation of plasmacytomasa with the chromosomal translocation t(12; 15) in interleukin 6 transgenic mice. Proc Natl Acad Sci USA 1992; 89: 232-235. PMID: 1729694
42. Wu G, Chai N, Kim I, Klein AS, Jordan SC. Monoclonal anti-interleukin-6 receptor antibody attenuates donor-specific antibody responses in a mouse model of allosensitization. Transpl Immunol 2013; 28: 138-143. PMID: 23562586
43. Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood 2008; 112: 3959-3964. PMID: 18784373
44. Vo AA, Choi J, Kim I, et al. A phase I/II trial of the interleukin-6 receptor specific humanized monoclonal (Tocilizumab) + intravenous immunoglobulin in difficult to desensitize patients. Transplantation 2015; 99: 2356-2363. PMID 26018350
45. Zhang ZX, Huang X, Jiang J, et al. Natural killer cells play a critical role in allograft vasculopathy in an interleukin-6-dependent manner. Transplantation 2014; 98: 1029-1039. PMID: 25286056
46. Kang S, Tanaka T, Kishimoto T. Therapeutic uses of anti-interleuking-6 receptor antibody. Int Immunol 2015; 27: 21-29. PMID: 25142313
47. Aoyama A, Tonsho M, Ng CY, et al. Long-term lung transplantation in nonhuman primates. Am J Transplant 2015; 15: 1415-1420. PMID: 25772308
48. Ma M, Sun Q, Li X, et al. Blockade of IL-6/IL-6R signaling attenuates acute antibody-mediated rejection in a mouse cardiac transplantation model. Front Immunol 2021; 12: 778539. PMID: 34777394 49. Cabezas L, Jouve T, Malvezzi P, et al. Tocilizumab and active antibody-mediated rejection in kidney transplantation: A literature review. Front Immunol 2022; 13: 839380. PMID: 35493469
50. Choi J, Aubert O, Vo A, et al. Assessment of tocilizumab (anti-interleukin-6 receptor monoclonal) as a potential treatment for chronic antibody-mediated rejection and transplant glomerulopathy in HLA-sensitized renal allograft recipients. Am J Transplant 2017; 17: 2381-2389. PMID: 28199785
51. Lavacca A, Presta R, Gai C, et al. Early effects of first-line treatment with anti- interleukin-6 receptor antibody tocilizumab for chronic active antibody-mediated rejection in kidney transplantation. Clin Transplant 2020; 34: el3908. PMID: 32415711
52. Noble J, Giovannini D, Laamech R, et al. Tocilizumab in the treatment of chronic antibody-mediated rejection post kidney transplantation: Clinical and histological monitoring. Front Med 2021; 8: 790547. PMID: 35004757
53. Kumar D, Yakubu I, Safavi F, et al. Lack of histological and molecular signature response to tocilizumab in kidney transplants with chronic active antibody mediated rejection: A case series. Kidney360 2020; 1 : 663-670. PMID: 35372943
54. Massat M, Congy-Jolivet N, Hebral A, et al. Do anti-IL6R blockers have a beneficial effect in the treatment of antibody-mediated rejection resistant to standard therapy after kidney transplantation? Am J Transplant 2021; 21 : 1641-1649. PMID: 33141487
55. Jordan SC, Ammerman N, Choi J, et al. Evaluation of clazakizumab (anti- interleukin-6) in patients with treatment-resistant chronic active antibody-mediated rejection of kidney allografts. Kidney Int Rep 2022; 7: 720-731. PMID: 35497778
56. Doberer K, Duerr M, Halloran PF, et al. A randomized clinical trial of anti-IL-6 antibody clazakizumab in late antibody-mediated kidney transplant rejection. JASN 2021; 32: 708-722. PMID: 33443079
57. Moreno I, Vicente R, Ramos F, Vicente JL, Barbera M. Determination of interleukin-6 in lung transplantation: Association with primary graft dysfunction. Transplant Proc 2007; 39: 2425- 2426. PMID: 17889209
58. Moreno I, Mir A, Vicente R, et al. Analysis of interleuking-6 and interleukin-8 in lung transplantation : Correlation with nitric oxide administration. Transplant Proc 2008; 40: 3082- 3084. PMID: 19010201
59. Verleden SE, Martens A, Ordies S, et al. Immediate post-operative bronchoalveolar lavage IL-6 and IL-8 are associated with early outcomes after lung transplantation. Clin Transplant 2018; 32: el3219. PMID: 29405435 60. Humbert M, Delattre RM, Fattal S, et al. In situ production of interleukin-6 within human lung allografts displaying rejection or cytomegalovirus pneumonia. Transplantation 1993; 56: 623-627. PMID: 8212159
61. Husain S, Resende MR, Rajwans N, et al. Elevated CXCL10 (IP-10) in bronchoalveolar lavage fluid is associated with acute cellular rejection after human lung transplantation. Transplantation 2014; 97: 90-97. PMID: 24025324
62. Kurzrock R, Voorhees PM, Casper C, et al. A phase I, open-label study of Siltuximab, an anti- IL-6 monoclonal antibody, in patients with B-cell Non-Hodgkin Lymphoma, multiple myeloma, or Castleman disease. Clin Cancer Re 2013; 19: 3659- 3670. PMID: 23659971
63. January SE, Fester KA, Halverson LP, et al. Tocilizumab for antibody-mediated rejection treatment in lung transplantation. J Heart Lung Transplant 2023; 42: 1353- 1357. PMID: 37268051
64. Choy EH, De Benedetti F, Takeuchi T, Hashizume M, John MR, Kishimoto T. Translating IL-6 biology into effective treatments. Nat Rev Rheum 2020; 16: 335-345. PMID: 32327746
65. Nishimoto N, Terao K, Mima T, Nakahara H, Takagi N, Kakehi T. Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease. Blood 2008; 112: 3959-3964. PMID: 18784373
66. Garbers C, Heink S, Korn T, Rose-John S. Interleukin-6: Designing specific therapeutics for a complex cytokine. Nat Rev Drug Discov 2018; 17: 395-412. PMID: 29725131
67. Grabers C, Spudy B, Aparicio-Siegmund S, et al. An interleukin-6 receptordependent molecular switch mediates signal transduction of the IL-27 cytokine subunit p28 (IL-30) via a gpl30 protein receptor homodimer. J Biol Chem 2013; 288: 4346- 4354. PMID: 23209286
68. Rossi JF, Chiang HC, Lu ZY, et al. Optimisation of anti-interleukin-6 therapy: Precision medicine through mathematical modelling. Front Immunol 2022; 13: 919489. PMID: 35928820
69. Hudes G, Tagawa ST, Whang YE, et al. A phase 1 study of a chimeric monoclonal antibody against interleukin-6, siltuximab, combined with docetaxel in patients with metastatic castration-resistant prostate cancer. Invest New Drugs 2013; 31: 669-676. PMID: 22828917
70. Angevin E, Tabernero J, Elez E, et al. A phase I/II, multiple-dose, doseescalation study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with advanced solid tumors. Clin Cancer Res 2014; 20: 2192-2204. PMID: 24563479 71. Suzuki K, Ogura M, Abe Y, et al. Phase 1 study in Japan of siltuximab, an anti- IL-6 monoclonal antibody, in relapsed/refractory multiple myeloma. Int J Hematol 2015; 101 : 286-294. PMID: 25655379
72. Van Rhee F, Wong RS, Munshi N, et al. Siltuximab for multicentric Castleman's disease: A randomised, double-blind, placebo-controlled trial. Lancet Oncol 2014; 15: 966-974. PMID: 25042199
73. Van Rhee F, Casper C, Voorhees PM, et al. A phase 2, open-label, multicenter study of the long-term safety of siltuximab (an anti-interleuking-6 monoclonal antibody) in patients with multicentric Castleman disease. Oncotarget 2015; 6: 30408- 30419. PMID: 26327301
74. Van Rhee F, Casper C, Voorhees PM, et al. Long-term safety of siltuximab in patients with idiopathic multicentric Castleman disease: A presepecified, open-label, extension analysis of two trials. Lancet Haematol 2020; 7: e209-e217. PMID: 32027862
75. Davis CC, Shah KS, Lechowicz MJ. Clinical development of siltuximab. Curr Oncol Rep 2015; 17: 29. PMID: 25986720
76. Mayer CL, Xie L, Bandekar R, et al. Dose selection of siltuximab for multicentric Castleman’ s disease. Cancer Chemother Pharmacol 2015; 75: 1037-1045. PMID: 25784388
77. Heinrich PC, Castell JV, Andus T. Interleukin-6 and the acute phase response. Biochem J 1990; 265: 621-636. PMID: 1689567
78. Van Rhee F, Voorhees P, Dispenzieri A, et al. International, evidence-based consensus treatment guidelines for idiopathic multicentric Castleman disease. Blood 2018; 132: 2115- 2124. PMID: 30181172
79. Narkhede M, Si Stasi A, Bal S, Hardwick P, Giri S, Mehta A. Interim analysis of investigator-initiated phase 2 trial of Siltuximab in treatment of cytokine release syndrome and immune effector cell associated neurotoxicity related to CAR T-cell therapy. Transplant Cell Ther 2023; 29 (Supple): S133-134.
80. Yu C, Cao J, Wang L, Yang Y, Ni Y, Wang J. Measuring the bioactivity of anti-IL- 6/anti-IL-6R therapeutic antibodies: Presentation of a robust reporter gene assay. Anal Bioanal Chem 2018; 410: 7067-7075. PMID: 30178083
81. Carpagnano GE, Kharitonov SA, Foschino-Barbaro MP, Resta O, Gramiccioni E, Barnes PJ. Increased inflammatory markers in the exhaled breath condensate of cigarette smokers. Eur Respir J 2003; 21 : 589-593. PMID: 12762340
82. Bucchioni E, Kharitonov SA, Allegra L, Barnes PJ. High levels of interleukin-6 in the exhaled breath condensate of patients with COPD. Respir Med 2003; 97: 1299- 1302. PMID: 14682411 83. Kumar R, Ison MG. Opportunistic infections in transplant patients. Infect Dis Clin N Am 2019; 33: 1143-1157. PMID: 31668195
84. Eur. J. Biochem (1987) 168, 543-550
85. J. Immunol. (1988)140, 1534-1541
86. Agr. Biol. Chem. (1990)54, 2685-2688
87. Chen F et al "Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CA T cell therapy" J. Immunol. Methods 2016, 434, 1-888. Brakenhoff, J. et al. (1990) J. Immunology 145: 561-568
89. "Monoclonal Antibodies; A manual of techniques", H Zola (CRC Press, 1988)
90. "Monoclonal Hybridoma Antibodies: Techniques and Application", SGR Hurrell (CRC Press, 1982)
91. Lee, E., Liang, Q., Ali, H. et al. Complete humanization of the mouse immunoglobulin loci enables efficient therapeutic antibody discovery. Nat Biotechnol 32, 356-363 (2014). https://doi.org/10.1038/nbt.2825
92. Neuberger et al (1998, 8th International Biotechnology Symposium Part 2, 792- 799)
93. Shaw, S., Bourne, T., Meier, C., Carrington, B., Gelinas, R., & Henry, A., et al., (2014), Discovery and characterization of olokizumab, mAbs, 6(3), 773-781
94. Wijdenes J, Clement C, Klein B, et al., Human recombinant dimeric IL-6 binds to its receptor as detected by anti-IL-6 monoclonal antibodies, Mol Immunol. 1991;28(11): 1183-1192
95. Fulciniti, M., Hideshima, T., Vermot-Desroches, C., Pozzi, S., Nanjappa, P., Shen, Z.,. & Tai, Y. T., (2009), A high-affinity fully human anti-IL-6 mAb, 1339, for the treatment of multiple myeloma, Clinical Cancer Research, 15(23), 7144-7152
96. Mease PJ, Gottlieb AB, et al. (September 2016). "The efficacy and safety of clazakizumab, an anti-interleukin-6 monoclonal antibody, in a phase lib study of adults with active psoriatic arthritis". Arthritis Rheumatol. 68 (9): 2163-73
97. Smolen JS, Weinblatt ME, Sheng S, Zhuang Y, Hsu B. Sirukumab, a human anti- interleukin-6 monoclonal antibody: a randomised, 2-part (proof-of-concept and dosefinding), phase II study in patients with active rheumatoid arthritis despite methotrexate therapy. Ann Rheum Dis. 2014 Sep;73(9): 1616-25. doi:
10.1136/annrheumdis-2013-205137. Epub 2014 Apr 3. PMID: 24699939; PMCID: PMC4145446
98. Matsuda, T. et al., Eur. J. Immunol. (1988) 18, 951-956
99. Sato, K. et al., The abstracts of the 21st Annual Meeting of the Japanese Society for Immunology (1991) 21, 166
100. Levilimab, Wikipedia entry, "https://en.wikipedia.org/wiki/Levilimab" accessed 19 April 2024 APPENDIX 1
Table 5 - Cohort A Table 6 - Cohort A2
Table 7 - Cohort B
Table 8 - Cohort C
Table 9 - Cohort D APPENDIX 2
X* indicates 2 measurements of pharmacokinetics on days that an investigational agent dose is given: a pre-dose measurement and a postdose measurement 1 hour after completing the infusion.

Claims

1. A method of treatment of antibody mediated rejection (AMR) of a lung transplant in a subject in need thereof, the method comprising a combination therapy of:
(i) administration to the subject of an antibody or fragment which is capable of inhibiting human IL-6; and
(ii) administration to the subject of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
2. The method according to Claim 1, wherein the subject is a human.
3. The method according to Claim 1 or 2, wherein the method is initiated after the transplant of a lung from a donor.
4. The method according to any one of Claims 1-3, wherein the lung transplant is a single or bilateral lung transplant.
5. The method according to any one of Claims 1-4, wherein the antibody or fragment which is capable of inhibiting human IL-6 is a chimeric, humanized or CDR grafted antibody or fragment thereof.
6. The method according to any one of Claims 1-5, wherein the antibody or fragment which is capable of inhibiting human IL-6 is a chimeric, humanized or CDR grafted antibody or fragment thereof comprising a heavy chain variable region in which CDR1, CDR2 and CDR3 comprise the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2 and SEQ ID NO: 3, respectively; and a light chain variable region in which CDR1, CDR2 and CDR3 comprise the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5 and SEQ ID NO: 6, respectively; and a constant region derived from a human IgG antibody.
7. The method according to any one of Claims 1-6, wherein the antibody or fragment which is capable of inhibiting human IL-6 is selected from siltuximab, olokizumab, elsilimomab, mAb 1339, clazakizumab, sirukumab, levilimab, SK2, MH166, and ARGX-109.
8. The method according to any one of Claims 1-7, wherein the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab.
9. The method according to any one of Claims 1-8, wherein the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is in a dose of 1 mg/kg to 15 mg/kg, optionally wherein the dose is 5.5 mg/kg or 11 mg/kg.
10. The method according to Claim 9, wherein the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab, and is administered in a dose of 5.5 mg/kg or 11 mg/kg.
11. The method according to any one of Claims 8-10, wherein the siltuximab is administered as 2 or more doses, optionally as 3 doses.
12. The method according to Claim 11, wherein a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 11 mg/kg on day 8 or day 22 of treatment.
13. The method according to Claim 11, wherein a first dose of siltuximab is administered at 11 mg/kg on day 1 of treatment, a second dose of siltuximab is administered at 11 mg/kg on day 8 of treatment, and a third dose of siltuximab is administered at 11 mg/kg on day 22 of treatment.
14. The method according to Claim 11, wherein a first dose of siltuximab is administered at 5.5 mg/kg on day 1 of treatment, and a second dose of siltuximab is administered at 5.5 mg/kg on day 8 or day 22 of treatment, optionally wherein the second dose is on day 8 of treatment and a third dose of siltuximab is administered at 5.5 mg/kg on day 22 of treatment.
15. The method according to any one of Claims 1-14, wherein the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is by intravenous administration, optionally by infusion, optionally wherein the infusion is over the course of one hour.
16. The method according to any one of Claims 1-15, wherein the AMR comprises the presence of donor-specific antibodies to human leukocyte antigens in the subject and/or wherein the AMR comprises allograft dysfunction.
17. The method according to any one of Claims 1-16, wherein the AMR is probable AMR or definite AMR.
18. The method according to any one of Claims 1-17, wherein the administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6 is given in a first treatment dose initiated within the period 24 hours before or after initiation of the immunosuppressive therapy, optionally wherein administration of the first treatment dose is initiated within the period 2 hours before or after initiation of the immunosuppressive therapy, optionally wherein administration of the first treatment dose is initiated within the period one hour before or after initiation of the immunosuppressive therapy, optionally wherein administration of the first treatment dose is initiated substantially at the same time as initiation of the immunosuppressive therapy.
19. The method according to any one of Claims 1-18, wherein the immunosuppressive therapy is administered 30 to 60 minutes prior to an or each administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6; optionally wherein the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab.
20. The method according to any one of Claims 1-19, wherein the immunosuppressive therapy comprises a multi-drug regimen.
21. The method according to any one of Claims 1-20, wherein the immunosuppressive therapy comprises administering one or more of a baseline immunosuppression maintenance therapy, and/or a standard of care treatment for AMR.
22. The method according to Claim 21, wherein the immunosuppressive therapy comprises a standard of care treatment for AMR comprising one or more of a corticosteroid, intravenous immune globulin (IVIG), Rituximab, Bortezomib, Carfilzomib, anti-thymocyte globulin (ATG), plasma exchange (PLEX), tacrolimus, cyclosporine A, Mycophenolate Mofetil (MMF), azathioprine, prednisone, and methylprednisolone, and optionally acetaminophen (paracetamol) and/or diphenhydramine.
23. The method according to Claim 22, wherein the corticosteroid is selected from one or more of prednisolone, prednisone, methylprednisolone, cortisone, dexamethasone, betamethasone and hydrocortisone.
24. The method according to any one of Claims 20-23, wherein the immunosuppressive therapy comprises acetaminophen, diphenhydramine, and methylprednisolone; optionally wherein the acetaminophen, diphenhydramine, and methylprednisolone are administered 30-60 minutes prior to an or each administration to the subject of the antibody or fragment which is capable of inhibiting human IL-6; optionally wherein the antibody or fragment which is capable of inhibiting human IL-6 is siltuximab.
25. The method according to Claim 24, wherein the immunosuppressive therapy comprises administering acetaminophen 650 or 1000 mg by mouth, diphenhydramine 25 or 50 mg by mouth, and methylprednisolone 60 mg intravenously, and wherein the administration of the antibody or fragment which is capable of inhibiting human IL-6 is administration of siltuximab; optionally wherein the acetaminophen, diphenhydramine, and methylprednisolone are administered 30-60 minutes prior to an or each administration of the siltuximab.
26. The method according to any one of Claims 1-25, wherein the method further comprises administration to the subject of a prophylactic medication, wherein the prophylactic medication is an antiviral and/or antimicrobial medication, optionally wherein the prophylactic medication comprises one or more of Valganciclovir, Trimethoprim-Sulfamethoxazole, Voriconazole, Posaconazole, and Isavuconazonium.
27. The method according to any one of Claims 21-26, wherein the baseline immunosuppression maintenance therapy comprises one or more of tacrolimus, cyclosporine A, Mycophenolate Mofetil (MMF), azathioprine, and prednisone.
28. The method according to Claim 27, wherein the baseline immunosuppression maintenance therapy comprises:
(a) tacrolimus, or cyclosporine A;
(b) Mycophenolate Mofetil (MMF), or azathioprine; and
(c) optionally prednisone.
29. The method according to any one of Claims 1-28, wherein the administration of the combination therapy reduces one or more of the severity of allograft dysfunction, the severity of abnormal lung histology, and the amount of donor-specific antibodies in the subject.
30. An anti-IL-6 antibody or fragment for use in treatment of an antibody mediated rejection (AMR) of a lung transplant, wherein the treatment comprises a combination therapy of administration of the antibody or fragment and administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
31. Use of an anti-IL-6 antibody or fragment in treatment of antibody mediated rejection (AMR) of a lung transplant, wherein the treatment comprises a combination therapy of administration of the antibody or fragment and administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL-6.
32. Use of an anti-IL-6 antibody or fragment for the manufacture of a medicament for treatment of antibody mediated rejection (AMR) of a lung transplant, wherein the medicament is to be administered in a combination therapy with administration of an immunosuppressive therapy, wherein the immunosuppressive therapy does not comprise any antibody or fragment thereof which is capable of inhibiting human IL- 6.
33. An anti-IL-6 antibody or fragment for use according to Claim 30, or use of an anti- IL-6 antibody or fragment according to Claim 31 or 32, having any of the additional features of any of Claims 1 to 29.
PCT/US2025/025686 2024-04-22 2025-04-22 Use of il-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject Pending WO2025226628A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202463637004P 2024-04-22 2024-04-22
US63/637,004 2024-04-22

Publications (1)

Publication Number Publication Date
WO2025226628A1 true WO2025226628A1 (en) 2025-10-30

Family

ID=95821552

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2025/025686 Pending WO2025226628A1 (en) 2024-04-22 2025-04-22 Use of il-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject

Country Status (1)

Country Link
WO (1) WO2025226628A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004039826A1 (en) 2001-11-14 2004-05-13 Centocor, Inc. Anti-il-6 antibodies, compositions, methods and uses
WO2010056948A2 (en) * 2008-11-13 2010-05-20 Femta Pharmaceuticals, Inc. Humanized anti-il-6 antibodies
WO2017106312A1 (en) * 2015-12-18 2017-06-22 Drexel University Methods of modulating levels of il-6 and pd-l1
US20210070853A1 (en) * 2018-01-04 2021-03-11 Vitaeris, Inc. Use of Anti-Il-6 Antibody, e.g., Clazakizumab for Desensitization of Solid Organ Transplant Recipients and/or for Preventing, Stabilizing or Reducing Antibody Mediated Rejection (ABMR)
WO2021046361A1 (en) * 2019-09-04 2021-03-11 Cedars-Sinai Medical Center Use of calcineurin inhibitor free ctla4-ig + anti-il6/il6r for long term immunosuppression in solid organ transplant recipients

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004039826A1 (en) 2001-11-14 2004-05-13 Centocor, Inc. Anti-il-6 antibodies, compositions, methods and uses
WO2010056948A2 (en) * 2008-11-13 2010-05-20 Femta Pharmaceuticals, Inc. Humanized anti-il-6 antibodies
WO2017106312A1 (en) * 2015-12-18 2017-06-22 Drexel University Methods of modulating levels of il-6 and pd-l1
US20210070853A1 (en) * 2018-01-04 2021-03-11 Vitaeris, Inc. Use of Anti-Il-6 Antibody, e.g., Clazakizumab for Desensitization of Solid Organ Transplant Recipients and/or for Preventing, Stabilizing or Reducing Antibody Mediated Rejection (ABMR)
WO2021046361A1 (en) * 2019-09-04 2021-03-11 Cedars-Sinai Medical Center Use of calcineurin inhibitor free ctla4-ig + anti-il6/il6r for long term immunosuppression in solid organ transplant recipients

Non-Patent Citations (99)

* Cited by examiner, † Cited by third party
Title
AGBOR-ENOH SJACKSON AMTUNC I ET AL.: "Late manifestation of alloantibody-associated injury and clinical pulmonary antibody-mediated rejection: Evidence from cell-free DNA analysis", J HEART LUNG TRANSPLANT, vol. 37, 2018, pages 925 - 932
AGR. BIOL. CHEM., vol. 54, 1990, pages 2685 - 2688
AGUILAR PRCARPENTER DRITTER J ET AL.: "The role of C4d deposition in the diagnosis of antibody-mediated rejection after lung transplantation", AM J TRANSPLANT, vol. 18, 2018, pages 936 - 944
ANGEVIN ETABERNERO JELEZ E ET AL.: "A phase I/II, multiple-dose, dose-escalation study of siltuximab, an anti-interleukin-6 monoclonal antibody, in patients with advanced solid tumors", CLIN CANCER RES, vol. 20, 2014, pages 2192 - 2204
AOYAMA ATONSHO MNG CY ET AL.: "Long-term lung transplantation in nonhuman primates", AM J TRANSPLANT, vol. 15, 2015, pages 1415 - 1420
BETTELLI ECARRIER YGAO W ET AL.: "Reciprocal developmental pathways for the generation of pathogenic effector Th17 and regulatory T cells", NATURE, vol. 441, 2006, pages 235 - 238, XP055049368, DOI: 10.1038/nature04753
BOS SMILROSS LFILBY AJVOS RFISHER AJ: "Immune processes in the pathogenesis of chronic lung allograft dysfunction: Identifying the missing pieces of the puzzle", EUR RESPIR REV, vol. 31, no. 165, 2022, pages 220060
BRAKENHOFF, J ET AL., J. IMMUNOLOGY, vol. 145, 1990, pages 561 - 568
BUCCHIONI EKHARITONOV SAALLEGRA LBARNES PJ: "High levels of interleukin-6 in the exhaled breath condensate of patients with COPD", RESPIR MED, vol. 97, 2003, pages 1299 - 1302
BURTON CMIVERSEN MCARLSEN J ET AL.: "Acute cellular rejection is a risk factor for bronchiolitis obliterans syndrome independent of post-transplant baseline FEV1", J HEART LUNG TRANSPLANT, vol. 28, 2009, pages 888 - 893, XP026519374, DOI: 10.1016/j.healun.2009.04.022
CABEZAS LJOUVE TMALVEZZI P ET AL.: "Tocilizumab and active antibody-mediated rejection in kidney transplantation: A literature review", FRONT IMMUNOL, vol. 13, 2022, pages 839380
CANTU EAPPEL JZHARTWIG MG ET AL.: "J. Maxwell Chamberlain Memorial Paper. Early fundoplication prevents chronic allograft dysfunction in patients with gastroesophageal reflux disease", ANN THORAC SURG, vol. 78, 2004, pages 1142 - 51
CARPAGNANO GEKHARITONOV SAFOSCHINO-BARBARO MPRESTA OGRAMICCIONI EBARNES PJ: "Increased inflammatory markers in the exhaled breath condensate of cigarette smokers", EUR RESPIR J, vol. 21, 2003, pages 589 - 593
CHAMBERS DC, CHERIKH WS, HARHAY MO: "The International Thoracic Organ Transplant Registry of the International Society for Heart and Lung Transplantation: Thirty-sixth adult lung and heart-lung transplantation report - 2019", DONOR AND RECIPIENT SIZE MATCH. J HEART LUNG TRANSPLANT, vol. 38, 2019, pages 1042 - 1055, XP085832282, DOI: 10.1016/j.healun.2019.08.001
CHEN F ET AL.: "Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CART cell therapy", J. IMMUNOL. METHODS, vol. 434, 2016, pages 1 - 888
CHOI JAUBERT OVO A ET AL.: "Assessment of tocilizumab (anti-interleukin-6 receptor monoclonal) as a potential treatment for chronic antibody-mediated rejection and transplant glomerulopathy in HLA-sensitized renal allograft recipients", AM J TRANSPLANT, vol. 17, 2017, pages 2381 - 2389, XP055707260, DOI: 10.1111/ajt.14228
CHOY EHDE BENEDETTI FTAKEUCHI THASHIZUME MJOHN MRKISHIMOTO T: "Translating IL-6 biology into effective treatments", NAT REV RHEUM, vol. 16, 2020, pages 335 - 345, XP038002564, DOI: 10.1038/s41584-020-0419-z
DAUD SAYUSEN RDMEYERS BF ET AL.: "Impact of immediate primary lung allograft dysfunction on bronchiolitis obliterans syndrome", AM J RESPIR CRIT CARE MED, vol. 175, 2007, pages 507 - 513
DAVIS CCSHAH KSLECHOWICZ MJ: "Clinical development of siltuximab", CURR ONCOL REP, vol. 17, 2015, pages 29, XP037917073, DOI: 10.1007/s11912-015-0453-1
DIEHL SASCHMIDLIN HNAGASAWA MBLOM BSPITS H: "IL-6 triggers IL-21 production by human CD4+ T cells to drive STAT3-dependent plasma cell differentiation in B cells", IMMUNOL CELL BIOL, vol. 90, 2012, pages 802 - 811
DOBERER KDUERR MHALLORAN PF ET AL.: "A randomized clinical trial of anti-IL-6 antibody clazakizumab in late antibody-mediated kidney transplant rejection", JASN, vol. 32, 2021, pages 708 - 722
DOBERER KONSTANTIN ET AL: "A Randomized Clinical Trial of Anti-IL-6 Antibody Clazakizumab in Late Antibody-Mediated Kidney Transplant Rejection", JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY, vol. 32, no. 3, 1 March 2021 (2021-03-01), pages 708 - 722, XP093295532, ISSN: 1046-6673, Retrieved from the Internet <URL:https://pmc.ncbi.nlm.nih.gov/articles/PMC7920172/> DOI: 10.1681/ASN.2020071106 *
DONG C: "Diversification of T-helper-cell lineages: Finding the family root of IL-17-producing cells", NAT REV IMMUNOL, vol. 6, 2006, pages 329 - 333
EUR. J. BIOCHEM, vol. 168, 1987, pages 543 - 550
FINLEN COPELAND CASNYDER LDZAAS DWTUBYFILL WJDAVIS WAPALMER SM: "Survival after bronchiolitis obliterans syndrome among bilateral lung transplant recipients", AM J RESPIR CRIT CARE, vol. 182, 2010, pages 784 - 9
FULCINITI, M.HIDESHIMA, T.VERMOT-DESROCHES, C.POZZI, S.NANJAPPA, P.SHEN, Z.TAI, Y. T.: "A high-affinity fully human anti-IL-6 mAb, 1339, for the treatment of multiple myeloma", CLINICAL CANCER RESEARCH, vol. 15, no. 23, 2009, pages 7144 - 7152, XP055045972, DOI: 10.1158/1078-0432.CCR-09-1483
GARBERS CHEINK SKORN TROSE-JOHN S: "Interleukin-6: Designing specific therapeutics for a complex cytokine", NAT REV DRUG DISCOV, vol. 17, 2018, pages 395 - 412, XP055753837, DOI: 10.1038/nrd.2018.45
GLANVILLE AR, VERLEDEN GM, TODD JL: "Chronic lung allograft dysfunction: Definition and update of restrictive allograft syndrome - A consensus report from the Pulmonary Council of the ISHLT", J HEART LUNG TRANSPLANT, vol. 38, 2019, pages 483 - 492
GLANVILLE ARABOYOUN CLHAVRYK A ET AL.: "Severity of lymphocytic bronchiolitis predicts long-term outcome after lung transplantation", AM J RESPIR CRIT CARE MED, vol. 177, 2008, pages 1033 - 1040
GOTTLIEB JVERLEDEN GMPERCH M ET AL.: "Disease progression in patients with the restrictive and mixed phenotype of chronic lung allograft dysfunction - A retrospective analysis in five European centers to assess the feasibility of a therapeutic trial", PLOS ONE, vol. 16, no. 12, 2021, pages e0260881
GRABERS CSPUDY BAPARICIO-SIEGMUND S ET AL.: "An interleukin-6 receptor-dependent molecular switch mediates signal transduction of the IL-27 cytokine subunit p28 (IL-30) via a gp130 protein receptor homodimer", J BIOL CHEM, vol. 288, 2013, pages 4346 - 4354
HEINK SYOGEV NGRABERS C ET AL.: "Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells", NAT IMMUNOL, vol. 18, 2017, pages 74 - 85, XP093097919, DOI: 10.1038/ni.3632
HEINRICH PCCASTELL JVANDUS T: "Interleukin-6 and the acute phase response", BIOCHEM J, vol. 265, 1990, pages 621 - 636
HIRANO TYASUKAWA KHARADA H ET AL.: "Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin", NATURE, vol. 324, 1986, pages 73 - 76, XP037103321, DOI: 10.1038/324073a0
HUANG HHYUSEN RDMEYERS BF ET AL.: "Late primary graft dysfunction after lung transplantation and bronchiolitis obliterans syndrome", AM J TRANSPLANT, vol. 8, 2008, pages 2454 - 2462, XP072338043, DOI: 10.1111/j.1600-6143.2008.02389.x
HUDES GTAGAWA STWHANG YE ET AL.: "A phase 1 study of a chimeric monoclonal antibody against interleukin-6, siltuximab, combined with docetaxel in patients with metastatic castration-resistant prostate cancer", INVEST NEW DRUGS, vol. 31, 2013, pages 669 - 676
HUMBERT MDELATTRE RMFATTAL S ET AL.: "In situ production of interleukin-6 within human lung allografts displaying rejection or cytomegalovirus pneumonia", TRANSPLANTATION, vol. 56, 1993, pages 623 - 627
HUSAIN SRESENDE MRRAJWANS N ET AL.: "Elevated CXCL10 (IP-10) in bronchoalveolar lavage fluid is associated with acute cellular rejection after human lung transplantation", TRANSPLANTATION, vol. 97, 2014, pages 90 - 97
J. IMMUNOL., vol. 140, 1988, pages 1534 - 1541
JANG MKTUNC IBERRY GJ ET AL.: "Donor -derived cell-free DNA accurately detects acute rejection in lung transplant patients, a multi-center cohort study", J HEART LUNG TRANSPLANT, vol. 40, 2021, pages 822 - 830
JANUARY SEFESTER KAHALVERSON LP ET AL.: "Tocilizumab for antibody-mediated rejection treatment in lung transplantation", J HEART LUNG TRANSPLANT, vol. 42, 2023, pages 1353 - 1357, XP087404944, DOI: 10.1016/j.healun.2023.05.012
JANUARY SPENSER E. ET AL: "Tocilizumab for antibody-mediated rejection treatment in lung transplantation", JOURNAL OF HEART AND LUNG TRANSPLANTATION, vol. 42, no. 10, 1 October 2023 (2023-10-01), AMSTERDAM, NL, pages 1353 - 1357, XP093295555, ISSN: 1053-2498, DOI: 10.1016/j.healun.2023.05.012 *
JORDAN SCAMMERMAN NCHOI J ET AL.: "Evaluation of clazakizumab (anti-interleukin-6) in patients with treatment-resistant chronic active antibody-mediated rejection of kidney allografts", KIDNEY INT REP, vol. 7, 2022, pages 720 - 731
KANG STANAKA TKISHIMOTO T: "Therapeutic uses of anti-interleuking-6 receptor antibody", INT IMMUNOL, vol. 27, 2015, pages 21 - 29, XP055377592, DOI: 10.1093/intimm/dxu081
KAWANO MHIRANO TMATSUDA T ET AL.: "Autocrine generation and requirement of BSF-2/IL-6 for human multiple myelomas", NATURE, vol. 332, 1988, pages 83 - 85, XP002199052, DOI: 10.1038/332083a0
KHALIFAH APHACHEM RRCHAKINALA MM ET AL.: "Respiratory viral infections are a distinct risk for bronchiolitis obliterans syndrome and death", AM J RESPIR CRIT CARE MED, vol. 170, 2004, pages 181 - 7
KISHIMOTO THIBI MMURAKAMI MNARAZAKI MSAITO MTAGA T: "The molecular biology of interleukin 6 and its receptor", CIBA FOUND SYMP, vol. 167, 1992, pages 5 - 16
KULKARNI HSCHERIKH WSCHAMBERS DC ET AL.: "Bronchiolitis obliterans syndrome-free survival after lung transplantation: An International Society for Heart and Lung Transplantation Thoracic Transplant Registry analysis", J HEART LUNG TRANSPLANT, vol. 38, 2019, pages 5 - 16, XP085559031, DOI: 10.1016/j.healun.2018.09.016
KUMAR DYAKUBU ISAFAVI F ET AL.: "Lack of histological and molecular signature response to tocilizumab in kidney transplants with chronic active antibody mediated rejection: A case series", KIDNEY360, vol. 1, 2020, pages 663 - 670
KUMAR RISON MG: "Opportunistic infections in transplant patients", INFECT DIS CLIN N AM, vol. 33, 2019, pages 1143 - 1157
KURZROCK RVOORHEES PMCASPER C ET AL.: "A phase I, open-label study of Siltuximab, an anti- IL-6 monoclonal antibody, in patients with B-cell Non-Hodgkin Lymphoma, multiple myeloma, or Castleman disease", CLIN CANCER RE, vol. 19, 2013, pages 3659 - 3670
LAVACCA APRESTA RGAI C ET AL.: "Early effects of first-line treatment with anti-interleukin-6 receptor antibody tocilizumab for chronic active antibody-mediated rejection in kidney transplantation", CLIN TRANSPLANT, vol. 34, 2020, pages e13908
LEE, E.LIANG, Q.ALI, H. ET AL.: "Complete humanization of the mouse immunoglobulin loci enables efficient therapeutic antibody discovery", NAT BIOTECHNOL, vol. 32, 2014, pages 356 - 363, Retrieved from the Internet <URL:https://doi.org/10.1038/nbt.2825>
LEVILIMAB, WIKIPEDIA ENTRY, 19 April 2024 (2024-04-19), Retrieved from the Internet <URL:https://en.wikipedia.org/wiki/Levilimab>
LEVINE DJGLANVILLE ARABOYOUN C ET AL.: "Antibody-mediated rejection of the lung: A consensus report of the International Society for Heart and Lung Transplantation", J HEART LUNG TRANSPLANT, vol. 35, 2016, pages 397 - 406, XP029492591, DOI: 10.1016/j.healun.2016.01.1223
LOBO LJARIS RMSCHMITZ JNEURINGER IP: "Donor-specific antibodies are associated with antibody-mediated rejection, acute cellular rejection, bronchiolitis obliterans syndrome, and cystic fibrosis after lung transplantation", J HEART LUNG TRANSPLANT, vol. 32, 2013, pages 70 - 77
MA CSDEENICK EKBATTEN MTANGYE SG: "The origins, function, and regulation of T follicular helper cells", J EXP MED, vol. 209, 2012, pages 1241 - 1253
MA MSUN QLI X ET AL.: "Blockade of IL-6/IL-6R signaling attenuates acute antibody-mediated rejection in a mouse cardiac transplantation model", FRONT IMMUNOL, vol. 12, 2021, pages 778539
MASSAT MCONGY-JOLIVET NHEBRAL A ET AL.: "Do anti-IL6R blockers have a beneficial effect in the treatment of antibody-mediated rejection resistant to standard therapy after kidney transplantation?", AM J TRANSPLANT, vol. 21, 2021, pages 1641 - 1649
MATSUDA, T ET AL., EUR. J. IMMUNOL., vol. 18, 1988, pages 951 - 956
MAYER CLXIE LBANDEKAR R ET AL.: "Dose selection of siltuximab for multicentric Castleman' s disease", CANCER CHEMOTHER PHARMACOL, vol. 75, 2015, pages 1037 - 1045, XP035497616, DOI: 10.1007/s00280-015-2720-0
MEASE PJGOTTLIEB AB ET AL.: "The efficacy and safety of clazakizumab, an anti-interleukin-6 monoclonal antibody, in a phase IIb study of adults with active psoriatic arthritis", ARTHRITIS RHEUMATOL., vol. 68, no. 9, September 2016 (2016-09-01), pages 2163 - 73, XP055846705, DOI: 10.1002/art.39700
MORENO IMIR AVICENTE R ET AL.: "Analysis of interleuking-6 and interleukin-8 in lung transplantation: Correlation with nitric oxide administration", TRANSPLANT PROC, vol. 40, 2008, pages 3082 - 3084
MORENO IVICENTE RRAMOS FVICENTE JLBARBERA M: "Determination of interleukin-6 in lung transplantation: Association with primary graft dysfunction", TRANSPLANT PROC, vol. 39, 2007, pages 2425 - 2426, XP022259738, DOI: 10.1016/j.transproceed.2007.07.056
MORRELL MRPILEWSKI JMGRIES CJ ET AL.: "De novo donor-specific HLA antibodies are associated with early and high-grade bronchiolitis obliterans syndrome and death after lung transplantation", J HEART LUNG TRANSPLANT, vol. 33, 2014, pages 1288 - 1294, XP029092358, DOI: 10.1016/j.healun.2014.07.018
NARKHEDE MSI STASI ABAL SHARDWICK PGIRI SMEHTA A: "Interim analysis of investigator-initiated phase 2 trial of Siltuximab in treatment of cytokine release syndrome and immune effector cell associated neurotoxicity related to CAR T-cell therapy", TRANSPLANT CELL THER, vol. 29, 2023, pages S133 - 134, XP087261526, DOI: 10.1016/S2666-6367(23)00234-8
NEUBERGER ET AL., 8TH INTERNATIONAL BIOTECHNOLOGY SYMPOSIUM, 1998, pages 792 - 799
NISHIMOTO NTERAO KMIMA TNAKAHARA HTAKAGI NKAKEHI T: "Mechanisms and pathologic significances in increase in serum interleukin-6 (IL-6) and soluble IL-6 receptor after administration of an anti-IL-6 receptor antibody, tocilizumab, in patients with rheumatoid arthritis and Castleman disease", BLOOD, vol. 112, 2008, pages 3959 - 3964, XP055046024, DOI: 10.1182/blood-2008-05-155846
NOBLE JGIOVANNINI DLAAMECH R ET AL.: "Tocilizumab in the treatment of chronic antibody-mediated rejection post kidney transplantation: Clinical and histological monitoring", FRONT MED, vol. 8, 2021, pages 790547
OTANI SDAVIS AKCANTWELL L ET AL.: "Evolving experience of treating antibody-mediated rejection following lung transplantation", TRANSPL IMMUNOL, vol. 31, 2014, pages 75 - 80
ROCHMAN IPAUL WEBEN-SASSON SZ: "IL-6 increases primed cell expansion and survival", J IMMUNOL, vol. 174, 2005, pages 4761 - 4767
ROSE-JOHN SNEURATH MF: "IL-6 trans-signaling: The heat is on", IMMUNITY, vol. 20, 2004, pages 2 - 4
ROSSI JF, CHIANG HC, LU ZY: "Optimisation of anti-interleukin-6 therapy: Precision medicine through mathematical modelling", FRONT IMMUNOL, vol. 13, 2022, pages 919489
ROUX ABENDIB LE LAN IHOLIFANJANIAINA S ET AL.: "Antibody-mediated rejection in lung transplantation: Clinical outcomes and donor-specific antibody characteristics", AM J TRANSPLANT, vol. 16, 2016, pages 1216 - 1228
ROYER P-JOLIVERA-BOTELLO GKOUTSOKERA A ET AL.: "Chronic lung allograft dysfunction: A systematic review of mechanisms", TRANSPLANTATION, vol. 100, 2016, pages 1803 - 1814
SAFAVI SROBINSON DRSORESI S ET AL.: "De novo donor HLA-specific antibodies predict development of bronchiolitis obliterans syndrome after lung transplantation", J HEART LUNG TRANSPLANT, vol. 33, 2014, pages 1273 - 1281, XP029092356, DOI: 10.1016/j.healun.2014.07.012
SATO MWADDELL TKWAGNETZ U ET AL.: "Restrictive allograft syndrome (RAS): A novel form of chronic lung allograft dysfunction", J HEART LUNG TRANSPLANT, vol. 30, 2011, pages 735 - 42, XP028226293, DOI: 10.1016/j.healun.2011.01.712
SATO, K. ET AL., THE ABSTRACTS OF THE 21ST ANNUAL MEETING OF THE JAPANESE SOCIETY FOR IMMUNOLOGY, vol. 21, 1991, pages 166
SCHAPER FROSE-JOHN S: "Interleukin-6: Biology, signaling and strategies of blockade", CYTOKINE GROWTH FACTOR REV, vol. 26, 2015, pages 475 - 487, XP029276459, DOI: 10.1016/j.cytogfr.2015.07.004
SGR HURRELL: "Monoclonal Hybridoma Antibodies: Techniques and Application", 1982, CRC PRESS
SHAW, S., BOURNE, T., MEIER, C., CARRINGTON, B., GELINAS, R., & HENRY, A.: "Discovery and characterization of olokizumab", MABS, vol. 6, no. 3, 2014, pages 773 - 781
SMOLEN JSWEINBLATT MESHENG SZHUANG YHSU B: "Sirukumab, a human anti-interleukin-6 monoclonal antibody: a randomised, 2-part (proof-of-concept and dose-finding), phase II study in patients with active rheumatoid arthritis despite methotrexate therapy", ANN RHEUM DIS., vol. 73, no. 9, 3 April 2014 (2014-04-03), pages 1616 - 25, XP055644870, DOI: 10.1136/annrheumdis-2013-205137
SUEMATSU SMATSUSAKA TMATSUDA T ET AL.: "Generation of plasmacytomasa with the chromosomal translocation t(12;15) in interleukin 6 transgenic mice", PROC NATL ACAD SCI USA, vol. 89, 1992, pages 232 - 235
SUZUKI KOGURA MABE Y ET AL.: "Phase 1 study in Japan of siltuximab, an anti-IL-6 monoclonal antibody, in relapsed/refractory multiple myeloma", INT J HEMATOL, vol. 101, 2015, pages 286 - 294, XP037917174, DOI: 10.1007/s12185-015-1743-y
TANAKA TKISHIMOTO T: "The biology and medical implications of interleukin-6", CANCER IMMUNOL RES, vol. 2, 2014, pages 288 - 294
TIKKANEN JMSINGER LGKIM SJ ET AL.: "De novo DQ donor-specific antibodies are associated with chronic lung allograft dysfunction after lung transplantation", AM J RESPIR CRIT CARE MED, vol. 194, 2016, pages 596 - 606
VAN RHEE FCASPER CVOORHEES PM ET AL.: "A phase 2, open-label, multicenter study of the long-term safety of siltuximab (an anti-interleuking-6 monoclonal antibody) in patients with multicentric Castleman disease", ONCOTARGET, vol. 6, 2015, pages 30408 - 30419
VAN RHEE FCASPER CVOORHEES PM ET AL.: "Long-term safety of siltuximab in patients with idiopathic multicentric Castleman disease: A presepecified, open-label, extension analysis of two trials", LANCET HAEMATOL, vol. 7, 2020, pages e209 - e217
VAN RHEE FVOORHEES PDISPENZIERI A ET AL.: "International, evidence-based consensus treatment guidelines for idiopathic multicentric Castleman disease", BLOOD, vol. 132, 2018, pages 2115 - 2124, XP055904003, DOI: 10.1182/blood-2018-07-862334
VAN RHEE FWONG RSMUNSHI N ET AL.: "Siltuximab for multicentric Castleman's disease: A randomised, double-blind, placebo-controlled trial", LANCET ONCOL, vol. 15, 2014, pages 966 - 974, XP055747111, DOI: 10.1016/S1470-2045(14)70319-5
VENADO AKUKREJA JGREENLAND JR: "Chronic lung allograft dysfunction", THORAC SURG CLIN, vol. 32, 2022, pages 231 - 242
VERLEDEN GM, GLANVILLE AR, LEASE ED: "Chronic lung allograft dysfunction: Definition, diagnostic criteria, and approaches to treatment - A consensus report from the Pulmonary Council of the ISHLT", J HEART LUNG TRANSPLANT, vol. 38, 2019, pages 493 - 503, XP055750689, DOI: 10.1016/j.healun.2019.03.009
VERLEDEN SEMARTENS AORDIES S ET AL.: "Immediate post-operative bronchoalveolar lavage IL-6 and IL-8 are associated with early outcomes after lung transplantation", CLIN TRANSPLANT, vol. 32, 2018, pages e13219
VO AACHOI JKIM I ET AL.: "A phase I/II trial of the interleukin-6 receptor specific humanized monoclonal (Tocilizumab) + intravenous immunoglobulin in difficult to desensitize patients", TRANSPLANTATION, vol. 99, 2015, pages 2356 - 2363, XP055809863, DOI: 10.1097/TP.0000000000000741
WIJDENES JCLEMENT CKLEIN B ET AL.: "Human recombinant dimeric IL-6 binds to its receptor as detected by anti-IL-6 monoclonal antibodies", MOL IMMUNOL, vol. 28, no. 11, 1991, pages 1183 - 1192, XP023852664, DOI: 10.1016/0161-5890(91)90004-4
WITT CAGAUT JPYUSEN RD ET AL.: "Acute antibody-mediated rejection after lung transplantation", J HEART LUNG TRANSPLANT, vol. 32, 2013, pages 1034 - 1040, XP028718214, DOI: 10.1016/j.healun.2013.07.004
WU GCHAI NKIM IKLEIN ASJORDAN SC: "Monoclonal anti-interleukin-6 receptor antibody attenuates donor-specific antibody responses in a mouse model of allosensitization", TRANSPL IMMUNOL, vol. 28, 2013, pages 138 - 143
YU CCAO JWANG LYANG YNI YWANG J: "Measuring the bioactivity of anti-IL-6/anti-IL-6R therapeutic antibodies: Presentation of a robust reporter gene assay", ANAL BIOANAL CHEM, vol. 410, 2018, pages 7067 - 7075, XP036614438, DOI: 10.1007/s00216-018-1307-0
ZHANG ZXHUANG XJIANG J ET AL.: "Natural killer cells play a critical role in allograft vasculopathy in an interleukin-6-dependent manner", TRANSPLANTATION, vol. 98, 2014, pages 1029 - 1039

Similar Documents

Publication Publication Date Title
US20240209102A1 (en) Methods of treating inflammatory conditions
KR101615420B1 (en) Diagnosis and treatments relating to th2 inhibition
US20210395376A1 (en) Antibodies and polypeptides directed against cd127
EP3111954B1 (en) Methods of treating ankylosing spondylitis using anti-il-17 antibodies
US8454965B2 (en) Method for the treatment of multiple sclerosis
CN104684931B (en) Anti-CD26 antibody and its use
US20210196822A1 (en) Treatment of triple negative breast cancer with targeted tgf-b inhibition
EP3626265A1 (en) Anti-human cd45rc antibodies and uses thereof
US20050089517A1 (en) Treatment of respiratory diseases with anti-IL-2 receptor antibodies
US20250313631A1 (en) Treatment and prevention of cancer using vista antigen-binding molecules
AU2024233551A1 (en) Methods for treating pulmonary fibrotic diseases or disorders with an anti oncostatin m receptor beta antibody
WO2025226628A1 (en) Use of il-6 inhibitors for preventing or inhibiting lung transplantation rejection in a subject
US20230272087A1 (en) Method of treating or preventing acute respiratory distress syndrome
WO2025224095A1 (en) Use of inhibitors of il-6 or il-6r for treating graft versus host disease in a subject
TW202436335A (en) Methods for treating dll3-expressing cancer
KR20230066320A (en) How to treat GD2 positive cancer
KR20230123477A (en) Methods for treating or preventing acute respiratory distress syndrome
HK1157192B (en) Method for the treatment of multiple sclerosis
HK1157192A (en) Method for the treatment of multiple sclerosis
HK1074769B (en) Method for the treatment of multiple sclerosis
HK1229257B (en) Methods of treating ankylosing spondylitis using anti-il-17 antibodies

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: 25727278

Country of ref document: EP

Kind code of ref document: A1