OBIVAS

A randomised, phase II, double blind, controlled mechanistic study of obinutuzumab versus rituximab in ANCA-associated vasculitis (ObiVas)

Research summary

ObiVas is a randomised, phase II, double blind, controlled trial designed to evaluate the mechanistic effect of obinutuzumab versus rituximab in active AAV. Participants will be randomised to one of two treatment groups in a 1:1 ratio and receive obinutuzumab (2 x 1000 mg, two weeks apart) plus prednisolone or rituximab (2 x 1000 mg, two weeks apart) plus prednisolone. All participants will undergo nasal biopsies at baseline and Week 26. Follow up will last for 18 months following entry into the study.

26 participants will be randomised and dosed with obinutuzumab/rituximab. Primary endpoint analysis will be performed after all participants have completed the 26 week biopsy and assessment.

6              Rationale for Trial

6.1          Rationale for the use of obinutuzumab: B cell depletion

B cells and T cells are both important in the autoimmune response in AAV. The central role of B cells is highlighted by the presence of activated B cells in inflammatory lesions, the association of B cell activation status with disease activity and the efficacy of B cell depletion therapy with rituximab (Voswinkel et al., 2008)(Popa et al., 1999)(Stone et al., 2010). The role of B cells extends beyond their function as precursors for ANCA-producing plasma cells. B cells can secrete pro-inflammatory cytokines, present antigen to autoreactive T cells and provide the co-stimulatory signals required for T cell activation.

ANCAs are usually of the immunoglobulin G (IgG) isotype, suggesting that the B cells that produce them have undergone class switching, a process that usually requires a germinal centre response and cognate T cell help (Brouwer et al., 1991). AAV is associated with an increased number of circulating CD4+ effector memory T cells, as well as with a functional defect in circulating regulatory T cells that may contribute to a breakdown in immune tolerance and to the emergence of pathogenic ANCA (Abdulahad et al. 2006)(Morgan et al., 2010)(Abdulahad et al. 2007). Furthermore, a disease-associated CD4 T peripheral helper subset has recently been defined in AAV, which displays cytotoxic potential and upregulated chemokine receptors that aid their migration to sites of inflammation. (Gopaluni, unpublished data). 

B cell depletion therapy with rituximab, a type I mAb directed against the B cell surface molecule CD20, is an effective strategy to both induce and maintain remission in AAV (Stone et al., 2010)(Jones et al., 2015). The cytotoxic effect is achieved by antibody-dependent cell-mediated cytotoxicity (ADCC), complement-mediated cell lysis and apoptosis. Although effective at inducing remission in patients with AAV, rituximab induction therapy does not improve subsequent relapse rates. When a single induction course of rituximab is used to treat a relapse, 75% of patients will relapse again within 2 years. Therefore, further doses of rituximab are needed to maintain remission, commonly delivered as fixed-interval (every 6 months), repeat doses over a 2-year period. However, there are complications related to repeated dosing of rituximab such as hypogammaglobulinaemia and increased susceptibility to infections. Thus, there is an unmet need to develop a treatment regimen that induces prolonged remission without a need for maintenance therapy.

After a single dose of rituximab, depletion of circulating peripheral blood B cells lasts for a median of 12 months (range 6-24), before B cells re-emerge at low level. Relapses usually coincide with the return of peripheral B cells (Stone et al. 2010); however, relapses can occur without detectable peripheral B cells, possibly driven by B cells in the tissue that are resistant to rituximab. The resistance of tissue B cells to depletion may reflect the limitations of the effector mechanisms of rituximab, or the presence of local B cell survival factors; indeed, increased levels of the B cell survival cytokine BAFF (B cell activating factor of the tumour necrosis factor family) observed following systemic B cell depletion may even enhance B cell survival within the tissue niche (Zhao et al. 2012). Furthermore, CD20-rituximab-complex internalisation and removal of CD20-rituximab by the monocytes from the cell surface (a process known as trogocytosis) may also reduce the half-life of rituximab (Golay et al., 2013). Thus, localised resistance to B cell depletion by rituximab may account for the treatment failures and inability of single courses of rituximab to induce long-lasting remission in patients with AAV.

Obinutuzumab is a type-II glycoengineered mAb directed against CD20 epitopes found only on B cells. Its modified structure leads to higher affinity CD20 antigen binding and increased direct cell death (DCD) compared to rituximab. It also enhances antibody-dependent cellular cytotoxicity (ADCC) by nearly 100-fold due to increased Fc binding affinity for activating Fc gamma receptors (FcyRs) on immune effector cells. It is not associated with surface CD20-mAb internalisation by B cells and is not susceptible to trogocytosis, all of which contribute to its superior potency compared to rituximab.

As discussed above, the efficacy of obinutuzumab and superior B cell depletion in comparison to rituximab has been shown in animal studies as well as in clinical trials involving patients with CLL and follicular lymphoma. Furthermore, in primates, depletion of B cells in the spleen and lymph nodes was greater with obinutuzumab compared to rituximab (Mössner et al., 2010).

The primary hypothesis for this trial is that B cell depletion in inflamed tissues in AAV patients is greater following obinutuzumab compared to rituximab. Secondary hypotheses are that greater tissue B cell depletion reduces B cell differentiation into ANCA-producing plasma cells and reduces antigen presentation by B cells to auto-reactive T cells, translating into more prolonged peripheral blood B cell depletion, greater normalisation of T cell repertoire with obinutuzumab versus rituximab, supporting the potential for longer treatment-free remissions with obinutuzumab.


Main inclusion criteria

Participants are eligible to be included in the trial only if all of the following criteria apply:

1.            Capable of giving signed informed consent.

2.            Participant must be >=18 years of age at the time of signing the informed consent form.

3.            Have a diagnosis of AAV (granulomatosis with polyangiitis or microscopic polyangiitis), according to the definitions of the Chapel Hill Consensus Conference (35).

4.            PR3 ANCA positivity by ELISA at screening.

5.            Have active disease defined by one major or three minor disease activity items on the Birmingham Vasculitis Activity Score for Wegener’s (BVAS/WG).

6.            Women of child-bearing potential (WOCBP) must agree to use effective contraception methods and agree to follow these methods for at least 18 months after the last dose of rituximab or obinutuzumab.

7.            Has received at least two doses of any COVID-19 vaccination

Main exclusion criteria

Exclusion criteria apply to the participant’s condition at screening unless otherwise stated.

Participants are excluded from the trial if any of the following criteria apply:

1.            Women who are pregnant, plan to become pregnant or breast feed during the trial.

2.            Current participation in any other interventional treatment trials.

3.            Compliance: is unlikely to comply with trial visits based on investigator judgment.

4.            MPO ANCA or anti–GBM antibody positivity by ELISA during screening.

5.            Presence of pulmonary haemorrhage with hypoxia.

6.            Estimated glomerular filtration rate (eGFR) <15 ml/min/1.73m2.

7.            Symptomatic herpes zoster within 3 months of screening.

8.            Evidence of active or latent tuberculosis (TB) determined by a positive (not indeterminate) QuantiFERON®-TB Gold test (or equivalent).

9.            Known hypersensitivity or significant allergies to monoclonal antibodies (including IMPs or to any of the excipients, e.g. murine proteins)

10.          Malignant neoplasm within 5 years (from screening) excluding basal cell or squamous cell carcinoma of the skin treated with local resection only or carcinoma in situ of the uterine cervix treated locally and without metastatic disease for 3 years.

11.          A history of a primary immunodeficiency or severe immunocompromise

12.          IgA deficiency (IgA < 10 mg/dL).

13.          IgG deficiency (IgG < 400 mg/dL).

14.          Neutrophils < 1.5 x 109 cells/L.

15.          B cell lymphopenia at screening (total CD19+ count <0.1x109/L).

16.          Alanine transferase (ALT) >2.5x upper limit of normal (ULN).

17.          Active bleeding disorders, and/or inability to support interruption to anticoagulant or anti-platelet therapies for nasal biopsy.

18.          Severe nasal deformity precluding endoscopic assessment/biopsy of postnasal space

19.          Severe heart failure (New York Heart Association Class IV) or other severe, uncontrolled cardiac disease.

20.          Have a history of a major organ transplant or hematopoietic stem cell/marrow transplant.

21.          Have an acute or chronic infection requiring management as follows:

• Currently on any treatment for a chronic infection such as pneumocystis, cytomegalovirus, herpes simplex virus, herpes zoster, or atypical mycobacteria

• Hospitalisation solely for treatment of proven infection requiring parenteral (IV or IM) antibiotics (antibacterials, antivirals, antifungals, or anti-parasitic agents) within 60 days of Day 1. NB Hospitalisation for a participant with active vasculitis with co-existent infection requiring IV or IM antibiotics is permitted. 

• Proven severe infection requiring outpatient treatment with parenteral (IV or IM) antibiotics (antibacterials, antivirals, antifungals, or anti-parasitic agents) within 60 days of Day 1. Prophylactic anti-infective treatment is allowed. Precautionary PO/IV antibiotics in a participant with active vasculitis is permitted.

22.          Positive human immunodeficiency virus (HIV) antibody test.

23.          Positive serology for Hepatitis B (HB), defined as: (i) HB surface antigen positive (HBsAg+) OR (ii) HB core antibody positive (HBcAb+)*.

24.          Positive Hepatitis C (HCV) antibody test.

25.          Any additional contraindication for IMP treatment as per the SmPCs

26.          Have clinical evidence of significant unstable or uncontrolled acute or chronic diseases not due to vasculitis which, in the opinion of the principal investigator, could confound the results of the trial or put the participant at undue risk.

27.          Have a planned surgical procedure, laboratory abnormality, or condition that, in the opinion of the principal investigator, makes the participant unsuitable for the trial.

28.          * Any positive hepatitis results will be reported to the appropriate authorities.

Prior/Concomitant Therapy:

29.          Live vaccine(s) within 30 days prior to Day 1, or plans to receive live vaccines during the trial.

30.          Have received any anti-CD20 (or any other B cell depleting therapies including alemtuzumab) within 12 months of Day 1.

31.          Have received any of the following within 180 days of Day 1:

• Cyclophosphamide

• Belimumab

32.          Have received any of the following within 90 days of Day 1:

• Anti-TNF or anti-IL-6 therapy (e.g., adalimumab, etanercept, infliximab, tocilizumab),

• Abatacept,

• Interleukin-1 receptor antagonist (e.g., anakinra),

• Intravenous immunoglobulin (IVIG),

• Plasmapheresis, leukapheresis.

33.          Have received any investigational agent (that is not approved for use in the UK) within 60 days of Day 1.

34.          Have received emergency IV steroid >3g methylprednisolone between 30 days prior to Screening Visit and up to Day 1 (including Day 1).


Funders and sponsors

Funders: MRC and Roche

Sponsors: CUH and University of Cambridge


Chief investigator

Dr Rachel Jones

Contact details

Clinical Trial Coordinator: Kim Mynard

Telephone: 01223 768317 | Email: [email protected]