Coverage Policy Manual
Policy #: 2021034
Category: Pharmacy
Initiated: November 2021
Last Review: October 2023
  Rituximab (e.g., Rituxan) and Biosimilars – Non-Oncologic Indications

Description:
Rituximab, an anti-CD20 monoclonal antibody, was originally approved by the FDA in November 1997 for the treatment of relapsed or refractory low grade or follicular CD20+ B-cell non-Hodgkin’s lymphoma (NHL).  Since its introduction there have been expanded FDA approved indications, and a number of off-label conditions for which the drug has been investigated.  
 
Rituximab has shown impressive benefit for some conditions, and in addition the drug has been found to have some serious adverse effects for which the FDA has issued "Black Box Warnings."  Fatal infusion reactions may occur within 24 hours of rituximab infusion; approximately 80% of fatal reactions occurred with first infusion. Monitor individuals and discontinue rituximab infusion for severe reactions. Acute renal failure requiring dialysis with instances of fatal outcome can occur in the setting of Tumor Lysis Syndrome (TLS) following treatment of rituximab in individuals with non-Hodgkin's lymphoma (NHL). Severe and potentially fatal mucocutaneous reactions can occur in individuals receiving rituximab. JC virus infection resulting in Progressive Multifocal Leukoencephalopathy (PML) and death can also occur in individuals receiving rituximab.
 
Regulatory Status
 
Rituximab and Hyaluronidase (e.g., Rituxan Hycela), is a combination of rituximab and hyaluronidase human, an endoglycosidase.  The FDA approved the rituximab and hyaluronidase, subcutaneous injection, in June 2017.  It is for the treatment of adult individuals with FL, CLL and DLBCL.  
 
On 11/28/2018, the FDA approved Rituximab abbs (e.g., Truxima) for the treatment of adult individuals with:
    1. Non-Hodgkin’s lymphoma (NHL)
    2. Relapsed or refractory, low grade or follicular, CD20-positive B-cell NHL as a single agent.
    3. Previously untreated follicular, CD20-positive, B-cell NHL in combination with first line chemotherapy and, in individuals achieving a complete or partial response to a rituximab product in combination with chemotherapy, as single-agent maintenance therapy.
    4. Non-progressing (including stable disease), low-grade, CD20 positive, B-cell NHL as a single agent after first-line cyclophosphamide, vincristine, and prednisone (CVP) chemotherapy.
 
On 5/23/19 the FDA approved biosimilar, Rituximab-abbs (e.g., Truxima), for the adult individuals with:
    1. Previously untreated diffuse Large B-cell, CD20-positive non-Hodgkin’s lymphoma (NHL) in combination with cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or other anthracycline-based chemotherapy regimens.
    2. Previously untreated and previously treated CD20-positive chronic lymphocytic leukemia (CLL) in combination with fludarabine and cyclophosphamide (FC).
 
On July 25, 2019, the Food and Drug Administration approved rituximab-pvvr (e.g., Ruxience), a biosimilar to Rituximab (e.g., Rituxan) for the treatment of adult individuals with non-Hodgkin’s lymphoma (NHL), chronic lymphocytic leukemia (CLL), and granulomatosis with polyangiitis (GPA) and microscopic polyangiitis (MPA).
 
On 12/18/2019 the FDA approved, biosimilar, Rituximab-abbs (e.g., Truxima) for the adult individuals with:
    1. Rheumatoid arthritis (RA) in combination with methotrexate in adult individuals with moderately-to severely active RA who have inadequate response to one or more TNF antagonist therapies.
    2. Granulomatosis with polyangiitis (GPA) Wegener's granulomatosis) and microscopic polyangiitis (MPA) in adult individuals win combination with glucocorticoids.
 
On December 17, 2020, the Food and Drug Administration approved rituximab-arrx (e.g., Riabni), the third biosimilar version of Rituxan for NHL, CLL, GPA, and MPA.
 
On June 3, 2022, the Food and Drug Administration approve rituximab-arrx (e.g., Riabni) for Rheumatoid Arthritis (RA) in combination with methotrexate in adult individuals with moderately -to-severely active RA who have inadequate response to one or more TNF antagonist therapies.
 
Coding
 
See CPT/HCPCS Code section below.

Policy/
Coverage:
For members of plans that utilize a prescription drug program vendor (i.e., Arkansas State Employees and Public School Employees, Arkansas State Police and Arkansas State University) for preferred products under the medical benefit, the preferred products contained in this policy are NOT applicable. Please contact the member’s prescription drug program vendor.
 
For coverage prior to November 1, 2021, please refer to policy number 2006016.
 
After November 1, 2021, please refer policy number 2006016 for oncological Rituximab uses.
 
Effective April 01, 2022, Prior Approval is required for rituximab.
 
The Step Therapy Medication Act is applicable to fully-insured (Arkansas Blue Cross, Health Advantage, and Exchange) and specified governmental (ASE/PSE and ASP) health plans. The law is not applicable to FEP or self-insured ERISA groups (including but not limited to Walmart or other Blue Advantage groups).   Initial approval for exigent request is 28 days. Otherwise, initial approval for standard review is up to 1 year.
 
Effective January 1, 2024
 
Select products (e.g., Truxima and Riabni) are preferred where there is an FDA approved indication for the biosimilar product and for all off-label uses of the reference product. According to the United States FDA “a biosimilar is a biological product that has no clinically meaningful differences from the existing FDA-approved reference product. All biosimilar products meet the FDA’s rigorous standards for approval for the indications described in the product labeling. Once a biosimilar has been approved by the FDA, the safety and effectiveness of these products have been established, just as they have been for the reference product.”
 
Preferred Products:
HCPCS                                                      Brand Name                                             Generic Name
Q5115                                                        Truxima                                                       Rituximab abbs
Q5123                                           Riabni                                            Rituximab arrx
 
Non-preferred Products:  
HCPCS                                                       Brand Name                                           Generic Name
J9310, J9312                                  Rituxan                                        Rituximab
J9311                                                          Rituxan Hycela                                      Rituximab and hyaluronidase
Q5119                                           Ruxience                                      Rituximab pvvr
 
If the request is for a non-preferred product, one of the following criteria must be met for the non-preferred product to be covered:
    1. The individual had a documented serious adverse event to all preferred products that required medical intervention AND the prescriber has completed and submitted an FDA MedWatch Adverse Event Reporting Form for each event (the prescriber must provide a copy of the completed MedWatch form. Authorizations will not be considered unless the form is completed and submitted to the FDA); OR
    2. None of the preferred products have an FDA approved indication that is requested, and the requested non-preferred product has the FDA approved indication that is requested.
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
  
Rituximab (e.g., Rituxan®) and biosimilars meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for the following indications:
 
FDA APPROVED INDICATIONS
 
RHEUMATOID ARTHRITIS
 
INITIAL APPROVAL STANDARD REVIEW for up to 12 months:
1. Individual is 18 years of age; AND
2.Individual has a diagnosis of moderate to severe rheumatoid arthritis (RA) supported by the submitted medical records; AND
3.Individual has an active disease with documented inadequate response (trial of 3 months) to at least one conventional synthetic DMARDs (e.g., methotrexate, sulfasalazine, leflunomide, hydroxychloroquine, cyclosporine) (Fraenkel, 2021); OR  
4.Individual has an active disease with documented intolerance or contraindication to at least one conventional synthetic DMARDs (e.g., methotrexate, sulfasalazine, leflunomide, hydroxychloroquine, cyclosporine) (Fraenkel, 2021); OR  
5.Individual has previously received a biologic (e.g., etanercept, infliximab, adalimumab, certolizumab, tocilizumab, sarilumab, golimumab, abatacept, anakinra, rituximab) or synthetic DMARD (tofacitinib, baricitinib, upadacitinib) indicated for rheumatoid arthritis (Fraenkel, 2021); AND
6.Individual is not using the medication in combination with other biologic intended for treatment of rheumatoid arthritis, including but not limited to: TNF inhibitor, IL-36 inhibitor, PDE4 inhibitor, any other IL inhibitor, or Janus kinase inhibitor; AND
7.Individual does not have latent tuberculosis or serious active infection; AND
8.Must be dosed in accordance with the FDA label.   
CONTINUED APPROVAL for up to 1 year:  
1.Individual has met criteria for initial approval; AND
2.Individual has experienced a documented positive clinical response; AND  
3.Individual is not using the medication in combination with other biologic intended for treatment of rheumatoid arthritis, including but not limited to: TNF inhibitor, IL-36 inhibitor, PDE4 inhibitor, any other IL inhibitor, or Janus kinase inhibitor; AND
4. Must be dosed in accordance with the FDA label.  
 
Anti-Neutrophil Cytoplasmic Antibody (ANCA) Vasculitides (associated small vessel vasculitis), Granulomatosis with Polyangiitis (GPA) (Wegener’s Granulomatosis) and Microscopic Polyangiitis (MPA)
 
INITIAL APPROVAL STANDARD REVIEW for up to 12 months:
1. Individual is 2 years of age and older; AND
2. Individual has had inadequate response or is intolerant to initial treatment with azathioprine, methotrexate, and/or mycophenolate.
 
CONTINUED APPROVAL for up to 1 year:  
1. Individual meets criteria for initial approval based on indication; AND
2. Individual has experienced a positive clinical response to rituximab; AND
3. Must be dosed in accordance with the FDA label unless otherwise specified.
 
Pemphigus Vulgaris (PV) and Pemphigus Foliaceus
 
INITIAL APPROVAL STANDARD REVIEW for up to 12 months:
1. Individual is unresponsive to conventional therapy (e.g., systemic corticosteroids and immunosuppressive agents); OR
2. Individual is intolerant to conventional therapy (e.g., systemic corticosteroids and immunosuppressive agents).
 
CONTINUED APPROVAL for up to 1 year:  
1. Individual meets criteria for initial approval based on indication; AND
2. Individual has experienced a positive clinical response to rituximab; AND
3. Must be dosed in accordance with the FDA label unless otherwise specified.
 
OFF-LABEL
 
INITIAL APPROVAL STANDARD REVIEW for up to 12 months:
 
1. Acquired inhibitors in Hemophilia – (Refractory or intolerant of cyclophosphamides and steroid therapy) [Collins 2009, Rossi 2016]
2. ANCA associated small vessel vasculitis: Churg-Strauss syndrome.
3. Autoimmune Hemolytic Anemia (AIHA) in individuals that have failed to respond to corticosteroid therapy [Shanafelt 2003, Birgens 2013, Michel 2017]
4. Autoimmune encephalitis (AE) when the following criteria are met [Graus, 2016]
a. Autoimmune limbic encephalitis - ALL criteria are required:
i. Subacute onset (rapid progression of < 3 months) of working memory deficits (short-term memory loss), seizures, or psychiatric symptoms suggesting involvement of the limbic system.
ii. Bilateral brain abnormalities on T2-weighted FLAIR MRI highly restricted to the medial temporal lobes
iii. At least one of the following:
1. CSF pleocytosis (> 5 white blood cells per mm to the 3rd power)
2. EEG with epileptic or slow-wave activity involving the temporal lobes.
b. Reasonable exclusion of alternative causes.
c. For continued therapy, diagnosis is confirmed by detection of a specific autoantibody associated with AE including but not limited to:
i. NMDAR, LGI1, Caspr2, AMPAR, GABA-A or GABA-B receptor, IgLON5, DPPX, GlyR, mGluR1, mGluR2, mGluR5, Neurexin 3-alpha, or dopamine-2 receptor (D2R)]; AND
ii. Individual has had an inadequate response to, is intolerant of, or has a contraindication to first line agent(s) including immunoglobulin therapy or plasma exchange.
5. Anti-NMDA receptor encephalitis (Graus, 2016) – ALL criteria below must be met:
a. Rapid onset (< 3 months) of at least four of the six following major groups of symptoms:
i. Abnormal (psychiatric) behavior or cognitive dysfunction
ii. Speech dysfunction
iii. Seizures
iv. Movement disorder, dyskinesias, or rigidity/abnormal postures
v. Decreased level of consciousness
vi. Autonomic dysfunction or central hypoventilation
b. At least one of the following laboratory results:
i. Abnormal EEG (focal or diffuse slow or disorganized activity, epileptic activity, or extreme delta brush), OR
ii. CSF with pleocytosis or oligoclonal bands
c. Reasonable exclusion of other disorders
d. For continuation of therapy,
i. IgG anti-GluN1 antibodies [diagnosis of anti-NMDA receptor encephalitis is confirmed by the detection of IgG antibodies to the GluN1 (also known as NR1) subunit of the NMDA receptor in serum or CSF] in the presence of one or more of the six major groups of symptoms, after reasonable exclusion of other disorders, AND
ii. Individual has had an inadequate response to, is intolerant of, or has a contraindication to first line agent(s) including immunoglobulin therapy or plasma exchange.
6. Catastrophic Antiphospholipid antibody Syndrome (CAPS) [Rao 2009]
7. Cryoglobulinemia associated with Sjogren’s or SLE [Ramos 2009]
8. Cryoglobulinemic vasculitis associated with Hepatitis C virus infection [Petrarca 2010, KDIGO 2018]
9. Ebstein-Barr Virus infection in individuals
10. With X-linked Immunodeficiency [Milone 2005]
11. Graft vs. Host Disease, Chronic [Cutler 2006]10.
12. Grave’s Ophthalmopathy [El Fassi 2007, Salvi 2007]
13. Immunoglobulin G4 related disease – (Failure or intolerance to steroid therapy.) [Carruthers 2015]
14. ITP, in children with ITP who have significant ongoing bleeding and/or have a need for improved quality of life despite conventional treatment. Also, may be considered as an alternative to splenectomy in children with chronic ITP or as therapy in those who have failed splenectomy. [Oved 2017
15. ITP, in adults at risk of bleeding who have failed one line of therapy such as corticosteroids, IVIg, or splenectomy. [Ghanima 2015]
16. Lupus Erythematosus, Systemic, as second- or third-line therapy [Merrill 2010]
17. Evan’s Syndrome, pediatric and adult, who are unresponsive to, or unable to tolerate corticosteroids [Bader-Meunier 2007, Rucker 2008]
18. Myasthenia Gravis generalized and MuSK positive Myasthenia Gravis. [Narayanaswami 2020, Singh 2019]
19. Membranous Glomerulonephropathy for individuals refractory to or cannot take alkylating agents or calcineurin inhibitors [Müller-Deile 2015, Anjum 2019].
20. Membranous Nephropathy (primary MN) In individuals with primary MN, initial therapy is based on an assessment of the individual’s risk of progressive disease [DeVriese, 2017]:
a. In individuals with primary MN regardless of phospholipase A2 receptor (PLA2R) – 2 or more of the following are considered very high risk for progressive disease:
b. Serum creatinine > 1.5 mg/dL [> 133 micromole/L] considered to be due to active MN.
c. Progressive decline in kidney function (e.g., decrease in estimated glomerular filtration rate [eGFR] > 25 percent from baseline over the prior two years) considered to be due to active MN.
d. Severe, disabling, or life-threatening nephrotic syndrome (defined by the presence of a serum albumin < 2.5 g/dL [if measured by bromocresol green methods] or < 2.0 g/dL [if measured by bromocresol purple methods] and refractory edema, or a thromboembolic event) For those individuals with primary MN and normal kidney function and do not have severe or life -threatening nephrotic syndrome, will observe for 3- 6 months (Including renin-angiotensin system inhibition). After 3-6 months of observation
 
High risk – Individuals with 2 or more of the following for progressive disease:
a. Decrease in eGFR of > 25 percent, not explained by other causes, at any time during the observation period.
b. Proteinuria > 8 g/day at the end of the observation period or persistent nephrotic syndrome (proteinuria > 3.5 g/day and serum albumin < 3.5 g/dL [if measured by bromocresol green methods] or < 3.0 g/dL [if measured by bromocresol purple methods])
c. If the individual is anti-PLA2R antibody positive, serial anti-PLA2R antibody titers are high (arbitrarily defined as > 150 RU/mL by the ELISA method) and not declining or are increasing to > 150 RU/mL.
 
Moderate risk – pts with 2 or more of the following for progressive disease:
a. Normal or stable eGFR (i.e., < 25 percent decrease) over the three-to-six-month period
b. Proteinuria < 4 g/day at the end of the observation period
c. If the individual is anti-PLA2R antibody positive, serial anti-PLA2R antibody titers are low (arbitrarily defined as < 50 RU/mL by the ELISA method) or decreasing by > 25 percent at three to six months.
21.Monoclonal Gammopathy of Unknown Significance with Polyneuropathy (with or without anti-MAG antibodies in individuals refractory to standard therapy [Chaudry 2017, Svahn, 2018]
22.Neuromyelitis Optica for individuals with disease refractory to immunosuppressive drugs and/or plasmapheresis [Nikoo, 2017]
23. Non-ANCA small vessel vasculitis: Henoch-Schönlein purpura and Cryoglobulinemic vasculitis
24. Multiple Sclerosis, Relapsing Remitting [Rae-Grant 2018]
25. Pediatric Nephrotic Syndrome –(Refractory or intolerant to steroid therapy and immunosuppressants.) [KDIGO 2012]
26. Rheumatoid arthritis, as a single agent in individuals who have failed DMARDs, and antitumor necrosis alfa drugs, and who have been shown to be intolerant of methotrexate FDA Approval
27. Sjögren’s Syndrome [Dass 2008]
28. Systemic Sclerosis (SSc)
a. Systemic Sclerosis (SSc)-associated Interstitial Lung Disease (ILD) (Goswami, 2021)
b. Previous trial/failure of standard treatment (i.e., Mycophenolate Mofetil (MMF), cyclophosphamide, or tocilizumab therapy).
c. Baseline pulmonary functional parameters [FVC (% predicted)]
d. Systemic Sclerosis (SSc)-Pulmonary Arterial Hypertension (PAH) (Zamanian, 2021)
e. Screening 6-minute Walking Distance (6MWD) of at least 100 meters
29. Thrombotic Thrombocytopenic Purpura, Acquired, due to ADAMTS13 deficiency - (refractory or relapsing to plasma exchange treatment and steroid therapy.) [Joly 2017]
30. Transplant, Heart, Acute Graft Rejection due to Antibody Mediated Disease, in individuals refractory to plasmapheresis/IVIG [Bierl 2006]
31. Transplant, Kidney, Desensitization of ABO Incompatible individuals on Renal Transplant Waiting List, when used in conjunction with IVIG [Echterdiek 2020, Wongsaroj 2015]
32. Transplant, Kidney, Desensitization of Panel Reactive Antibodies on Renal Transplant Waiting List [Vo 2010] Transplant, Kidney, Acute Graft Rejection due to Antibody Mediated Disease, when used as second-line therapy [KIDGO 2009]
33. Transplant, Lung, Acute Humoral Antibody Mediated Disease [Martinu 2010]
34. Transplant, Heart, Desensitization of Panel Reactive Antibodies in individuals on Cardiac Transplant Waiting List [Colvin, 2019]
 
Note: Please see non-covered indications and circumstances listed below.
 
CONTINUED APPROVAL for up to 1 year:  
 
1. Individual meets criteria for initial approval based on indication; AND
2. Individual has experienced a positive clinical response to rituximab.
 
Policy Guidelines  
 
Examples of Contraindications to Methotrexate:
 
1. Alcoholism, alcoholic liver disease or other chronic liver disease
2. Breastfeeding
3. Blood dyscrasias (e.g., thrombocytopenia, leukopenia, significant anemia)
4. Elevated liver transaminases
5. History of intolerance or adverse event
6. Hypersensitivity
7. Interstitial pneumonitis or clinically significant pulmonary fibrosis
8. Myelodysplasia
9. Pregnancy or planning pregnancy (male or female)
10. Renal impairment
11. Significant drug interaction
 
Dosage and Administration
Dosing per FDA Guidelines
 
RA
The dose for RA in combination with methotrexate is two-1,000 mg intravenous infusions separated by 2 weeks (one course) every 24 weeks or based on clinical evaluation, but not sooner than every 16 weeks. Methylprednisolone 100 mg intravenous or equivalent.
 
ANCA Vasculitides: GPA and MPA
Induction dose for adult individuals with active GPA and MPA in combination with glucocorticoids is 375 mg/m2 once weekly for 4 weeks. Follow-up dose for those who have achieved control with induction treatment, in combination with glucocorticoids is 500 mg/square meters IV separated by 2 weeks followed by 500 mg/square meters infusion every 6 months thereafter based on clinical evaluation.
 
Rituximab is available as 100 mg/10 mL (10 mg/mL) and 500 mg/50 mL (10 mg/mL) solution in single-dose vials.
 
Rituximab should be administered as an intravenous infusion by a healthcare professional.   
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
Rituximab and biosimilars, for any indication or circumstance not described above, do not meet member benefit Primary Coverage Criteria that there be scientific evidence of effectiveness in improving health outcomes for any other indications including the following conditions:
 
1. Atopic Eczema
2. Autoimmune Neutropenia
3. Autoimmune Retinopathy
4. Chronic Fatigue Syndrome
5. Chronic Inflammatory Demyelinating Polyneuropathy
6. Cryoglobulinemia for any indication except the use described under covered conditions.
7. Desensitization of HLA Incompatible individuals on liver transplant waiting list.
8. Desensitization of HLA Incompatible individuals on renal transplant waiting list when used as a single agent.
9. Ebstein-Barr Virus infection in any condition other than X-linked immunodeficiency
10. Glomerulosclerosis, Focal Segmental
11. Graft-Versus-Host Disease, Acute for treatment of active disease
12. Graft-Versus-Host Disease, Acute, as a preventative agent
13. Graft-Versus-Host Disease, Acute, for steroid-refractory disease
14. Graft-Versus-Host Disease, Chronic, as initial, (first line) therapy
15. Graft-Versus-Host Disease, Chronic, as preventative therapy
16. Fibrillary Glomerulonephritis· Hemophilia A, treatment to eradicate or suppress autoimmune anti-factor VIII antibodies.
17. IgA Nephropathy
18. Multiple Myeloma, for Primary, Recurrent, Relapsed, or Refractory Disease in individuals with absence of CD20+ antigen plasma cells
19. Multiple Sclerosis, Primary Progressive
20. Polymyositis
21. Pemphigus Vulgaris and Pemphigus Foliaceous, when administered concomitantly (or sequentially) with immune globulin.
22. Primary Biliary Cirrhosis
23. Red Cell Aplasia or Diamond Blackfan Anemia
24. Rheumatoid arthritis, when administered concomitantly (or within the same month) with anti-tumor necrosis alpha drugs.
25. Transplant, Heart, Desensitization of ABO Incompatible individuals on Transplant Waiting List
26. Transplant, Kidney, Desensitization of HLA Panel Reactive Antibodies developing in kidney transplant recipients to prevent potential rejection.
27. Transplant, Kidney, as Induction Given Prior to Transplant
28. Transplant, Liver, Desensitization of ABO Incompatible individuals on Transplant Waiting List
29. Transplant, Liver, Rejection
30. Transplant, Lung, Acute Rejection due to Cellular Vascular Rejection
31. Transplant, Lung, Desensitization of ABO Incompatible individuals on Transplant Waiting List
32. Urticaria, Chronic
 
Requests for coverage of any of the conditions on the non-Covered list, or for any condition not listed in the policy will require submission of data on effectiveness which will be reviewed to determine if the proposed indication meets member certificate of benefit coverage criteria.
 
For off-label use for non-malignant conditions, rituximab does not meet member certificate of benefit primary coverage criteria if:
 
1. The use is being studied in phase I, II, or III clinical trials or otherwise under study to determine effectiveness.
2. If there is lack of scientific evidence of effectiveness; or
3. The effectiveness of the particular use is in conflict or the subject of debate amongst experts.
 
For contracts without primary coverage criteria, any other use of rituximab that is not listed as a covered indication is investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective October 26, 2022 to December 31, 2023
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
1. RITUXIMAB (e.g., Rituxan®)
The use of Rituximab meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for the following indications:
 
INITIAL APPROVAL STANDARD REVIEW for up to 12 months:
 
FDA APPROVED INDICATIONS
        1. Rheumatoid Arthritis (RA), For those moderate to severely active, in combination with methotrexate for individuals who have had an inadequate response to one or more TNF antagonist therapies.
        2. Anti-Neutrophil Cytoplasmic Antibody (ANCA) Vasculitides (associated small vessel vasculitis), Granulomatosis with Polyangiitis (GPA) (Wegener’s Granulomatosis) and Microscopic Polyangiitis (MPA) in adult and pediatric individuals 2 years of age and older when initial treatment with azathioprine, methotrexate, and/or mycophenolate has been tried and failed or individual is intolerant.
        3. Pemphigus Vulgaris (PV) and Pemphigus Foliaceus, unresponsive to conventional therapy (e.g., systemic corticosteroids and immunosuppressive agents) or in those individuals in whom these drugs would not be tolerated.
 
CONTINUATION OF THERAPY for 12 months:
        1. Individual meets criteria for initial approval based on indication.
        2. Individual has experienced a positive clinical response to rituximab.
        3. Must be dosed in accordance with the FDA label unless otherwise specified.
 
Dosage and administration
 
Must be dosed in accordance with the FDA label unless otherwise specified.
 
RA
The dose for RA in combination with methotrexate is two-1,000mg IV infusions separated by 2 weeks, (one course) every 24 weeks, or based on clinical evaluation not sooner than every 16 weeks. Methylprednisolone 100 mg IV or equivalent glucocorticoid is recommended 30 min prior to each infusion.
 
ANCA Vasculitides: GPA and MPA
Induction dose for adult individuals with active GPA and MPA in combination with glucocorticoids is 375 mg/m2 once weekly for 4 weeks. Follow-up dose for those who have achieved control with induction treatment, in combination with glucocorticoids is 500 mg/m2 IV separated by 2 weeks followed by 500 mg/m2 infusion every 6 months thereafter based on clinical evaluation.
 
Induction dose for pediatric individuals in combination with glucocorticoids is 375 mg/m2 once weekly for 4 weeks. Follow-up dosing in combination with glucocorticoids is two 250 mg/m2 infusions separated by two weeks, followed by a 250 mg/m2 IV infusions every 6 months thereafter based on clinical evaluation.
 
PV
The dose for PV is two-1,000 mg IV infusion separated by 2 weeks in combination with a tapering course of glucocorticoids, then a 500 mg infusion at month 12 and every 6 months thereafter or based on clinical evaluation. Dose on relapse is a 1,000 mg infusion with considerations to resume or increase the glucocorticoid dose based on clinical evaluation. Subsequent infusion may be no sooner than 16 weeks after the previous infusion.
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
2. RITUXIMAB-PVVR (e.g., Ruxience), RITUXIMAB-ARRX (e.g., Riabni), and Rituximab-abbs (e.g., Truxima)
Rituximab-pvvr and Rituximab-arrx meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for the following indication:
 
FDA APPROVED INDICATIONS
        1. Anti-Neutrophil Cytoplasmic Antibody Vasculitis (ANCA) associated small vessel vasculitis, Granulomatosis with Polyangiitis (GPA) (Wegener’s Granulomatosis) and Microscopic Polyangiitis (MPA) in adult and pediatric individuals 2 years of age and older when initial treatment with azathioprine, methotrexate, and/or mycophenolate has been tried and failed or individual is intolerant.
        2. Rheumatoid Arthritis (RA) in combination with methotrexate in adult individuals with moderately-to severely active RA who have inadequate response to one or more TNF antagonist therapies.  
CONTINUATION OF THERAPY for 12 months:
        1. Individual meets criteria for initial approval based on indication.
        2. Individual has experienced a positive clinical response to rituximab.
        3. Must be dosed in accordance with FDA label unless otherwise specified.
 
Dosage and administration
 
Must be dosed in accordance with the FDA label unless otherwise specified.
 
ANCA Vasculitides: GPA and MPA
Induction dose for adult individuals with active GPA and MPA in combination with glucocorticoids is 375 mg/m2 once weekly for 4 weeks. Follow-up dose for those who have achieved control with induction treatment, in combination with glucocorticoids is 500 mg/m2 IV separated by 2 weeks followed by 500 mg/m2 infusion every 6 months thereafter based on clinical evaluation.
 
RA
The dose for RA in combination with methotrexate is two-1,000 mg intravenous infusions separated by 2 weeks (one course) every 24 weeks or based on clinical evaluation, but not sooner than every 16 weeks. Methylprednisolone 100 mg intravenous or equivalent.
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
OFF-LABEL INDICATIONS for Rituximab (e.g., Rituxan) and Biosimilars, (e.g., Ruxience; Truxima and Riabini)
The use of Rituximab and biosimilars meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for off-label use for treatment of the following conditions:
 
    1. Acquired inhibitors in Hemophilia – (Refractory or intolerant of cyclophosphamides and steroid therapy) [Collins 2009, Rossi 2016]
    2. ANCA associated small vessel vasculitis: Churg-Strauss syndrome
    3. Autoimmune Hemolytic Anemia (AIHA) in individuals that have failed to respond to corticosteroid therapy [Shanafelt 2003, Birgens 2013, Michel 2017]
    4. Autoimmune encephalitis (AE) when the following criteria are met [Graus, 2016]
·    Autoimmune limbic encephalitis - ALL criteria are required:
            • Subacute onset (rapid progression of <3 months) of working memory deficits (short-term memory loss), seizures, or psychiatric symptoms suggesting involvement of the limbic system
            • Bilateral brain abnormalities on T2-weighted FLAIR MRI highly restricted to the medial temporal lobes
            • At least one of the following:
· CSF pleocytosis (>5 white blood cells per mm3)
· EEG with epileptic or slow-wave activity involving the temporal lobes
·    Reasonable exclusion of alternative causes.
·    For continued therapy, diagnosis is confirmed by detection of a specific autoantibody associated with AE including but not limited to:
            • NMDAR, LGI1, Caspr2, AMPAR, GABA-A or GABA-B receptor, IgLON5, DPPX, GlyR, mGluR1, mGluR2, mGluR5, Neurexin 3-alpha, or dopamine-2 receptor (D2R)]; AND
            • individual has had an inadequate response to, is intolerant of, or has a contraindication to first line agent(s) including immunoglobulin therapy or plasma exchange
5. Anti-NMDA receptor encephalitis (Graus, 2016) – ALL criteria below must be met:
·   Rapid onset (<3 months) of at least four of the six following major groups of symptoms:
            • Abnormal (psychiatric) behavior or cognitive dysfunction
            • Speech dysfunction
            • Seizures
            • Movement disorder, dyskinesias, or rigidity/abnormal postures
            • Decreased level of consciousness
            • Autonomic dysfunction or central hypoventilation
·    At least one of the following laboratory results:
            • Abnormal EEG (focal or diffuse slow or disorganized activity, epileptic activity, or extreme delta brush), OR
            • CSF with pleocytosis or oligoclonal bands
·    Reasonable exclusion of other disorders
·    For continuation of therapy,
            • IgG anti-GluN1 antibodies [diagnosis of anti-NMDA receptor encephalitis is confirmed by the detection of IgG antibodies to the GluN1 (also known as NR1) subunit of the NMDA receptor in serum or CSF] in the presence of one or more of the six major groups of symptoms, after reasonable exclusion of other disorders, AND
            • individual has had an inadequate response to, is intolerant of, or has a contraindication to first line agent(s) including immunoglobulin therapy or plasma exchange
6. Catastrophic Antiphospholipid antibody Syndrome (CAPS) [Rao 2009]
7. Cryoglobulinemia associated with Sjogren’s or SLE [Ramos 2009]
8. Cryoglobulinemic vasculitis associated with Hepatitis C virus infection [Petrarca 2010, KDIGO 2018]
9. Ebstein-Barr Virus infection in individuals
10.  with X-linked Immunodeficiency [Milone 2005]
11. Graft vs. Host Disease, Chronic [Cutler 2006]10.
12. Grave’s Ophthalmopathy [El Fassi 2007, Salvi 2007]
13. Immunoglobulin G4 related disease – (Failure or intolerance to steroid therapy.) [Carruthers 2015]
14. ITP, in children with ITP who have significant ongoing bleeding and/or have a need for improved quality of life despite conventional treatment. Also, may be considered as an alternative to splenectomy in children with chronic ITP or as therapy in those who have failed splenectomy. [Oved 2017
15. ITP, in adults at risk of bleeding who have failed one line of therapy such as corticosteroids, IVIg, or splenectomy. [Ghanima 2015]
16. Lupus Erythematosus, Systemic, as second- or third-line therapy [Merrill 2010]
17. Evan’s Syndrome, pediatric and adult, who are unresponsive to, or unable to tolerate corticosteroids [Bader-Meunier 2007, Rucker 2008]
18. Myasthenia Gravis generalized and MuSK positive Myasthenia Gravis. [Narayanaswami 2020, Singh 2019]
19. Membranous Glomerulonephropathy for individuals refractory to, or cannot take alkylating agents or calcineurin inhibitors [Müller-Deile 2015, Anjum 2019]17
20. Membranous Nephropathy (primary MN) In individuals with primary MN, initial therapy is based on an assessment of the individual’s risk of progressive disease [DeVriese, 2017]:
          • In individuals with primary MN regardless of phospholipase A2 receptor (PLA2R) – 2 or more of the following are considered very high risk for progressive disease:
          • Serum creatinine 1.5 mg/dL [133 micromol/L] considered to be due to active MN
          • Progressive decline in kidney function (eg, decrease in estimated glomerular filtration rate [eGFR] 25 percent from baseline over the prior two years) considered to be due to active MN
          • Severe, disabling, or life-threatening nephrotic syndrome (defined by the presence of a serum albumin <2.5 g/dL [if measured by bromocresol green methods] or <2.0 g/dL [if measured by bromocresol purple methods] and refractory edema, or a thromboembolic event) For those individuals with primary MN and normal kidney function and do not have severe or life -threatening nephrotic syndrome, will observe for 3- 6 months (Including renin-angiotensin system inhibition). After 3-6 months of observation
 
High risk – Individuals with 2 or more of the following for progressive disease:
          • Decrease in eGFR of 25 percent, not explained by other causes, at any time during the observation period
          • Proteinuria >8 g/day at the end of the observation period or persistent nephrotic syndrome (proteinuria >3.5 g/day and serum albumin <3.5 g/dL [if measured by bromocresol green methods] or <3.0 g/dL [if measured by bromocresol purple methods])
          • If the individual is anti-PLA2R antibody positive, serial anti-PLA2R antibody titers are high (arbitrarily defined as 150 RU/mL by the ELISA method) and not declining or are increasing to 150 RU/mL
Moderate risk – pts with 2 or more of the following for progressive disease:
          • Normal or stable eGFR (ie, <25 percent decrease) over the three-to-six-month period
          • Proteinuria <4 g/day at the end of the observation period
          • If the patient is anti-PLA2R antibody positive, serial anti-PLA2R antibody titers are low (arbitrarily defined as <50 RU/mL by the ELISA method) or decreasing by 25 percent at three to six months
21. Monoclonal Gammopathy of Unknown Significance with Polyneuropathy (with or without anti-MAG antibodies in individuals refractory to standard therapy [Chaudry 2017, Svahn, 2018]
22. Neuromyelitis Optica for individuals with disease refractory to immunosuppressive drugs and/or plasmapheresis [Nikoo, 2017]
23. Non-ANCA small vessel vasculitis: Henoch-Schönlein purpura and Cryoglobulinemic vasculitis
24. Multiple Sclerosis, Relapsing Remitting [Rae-Grant 2018] Pediatric Nephrotic Syndrome –(Refractory or intolerant to steroid therapy and immunosuppressants.) [KDIGO 2012]
25. Rheumatoid arthritis, as a single agent in individuals who have failed DMARDs, and antitumor necrosis alfa drugs, and who have been shown to be intolerant of methotrexate FDA Approval
26. Sjögren’s Syndrome [Dass 2008]
27. Systemic Sclerosis (SSc)
          • Systemic Sclerosis (SSc)-associated Interstitial Lung Disease (ILD) (Goswami, 2021)
          • Previous trial/failure of standard treatment (i.e., Mycophenolate Mofetil (MMF), cyclophosphamide, or tocilizumab therapy).
          • Baseline pulmonary functional parameters [FVC (% predicted)]
          • Systemic Sclerosis (SSc)-Pulmonary Arterial Hypertension (PAH) (Zamanian, 2021)
          • Screening 6-minute Walking Distance (6MWD) of at least 100 meters
28.  Thrombotic Thrombocytopenic Purpura, Acquired, due to ADAMTS13 deficiency - (refractory or relapsing to plasma exchange treatment and steroid therapy.) [Joly 2017]
29.  Transplant, Heart, Acute Graft Rejection due to Antibody Mediated Disease, in individuals refractory to plasmapheresis/IVIG [Bierl 2006]
30.  Transplant, Kidney, Desensitization of ABO Incompatible individuals on Renal Transplant Waiting List, when used in conjunction with IVIG [Echterdiek 2020, Wongsaroj 2015]
31.  Transplant, Kidney, Desensitization of Panel Reactive Antibodies on Renal Transplant Waiting List [Vo 2010] Transplant, Kidney, Acute Graft Rejection due to Antibody Mediated Disease, when used as second-line therapy [KIDGO 2009]
32.  Transplant, Lung, Acute Humoral Antibody Mediated Disease [Martinu 2010]
33.  Transplant, Heart, Desensitization of Panel Reactive Antibodies in individuals on Cardiac Transplant Waiting List [Colvin, 2019]
 
CONTINUATION OF THERAPY for 12 months:
        1. Individual meets criteria for initial approval based on indication.
        2. Individual has experienced a positive clinical response to rituximab.
        3. Must be dosed in accordance with FDA label unless otherwise specified.
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
The use of Rituximab and biosimilars do not meet member benefit Primary Coverage Criteria that there be scientific evidence of effectiveness in improving health outcomes for any other indications including the following conditions:
 
    1. Atopic Eczema
    2. Autoimmune Neutropenia
    3. Autoimmune Retinopathy
    4. Chronic Fatigue Syndrome
    5. Chronic Inflammatory Demyelinating Polyneuropathy
    6. Cryoglobulinemia for any indication except the use described under covered conditions.
    7. Desensitization of HLA Incompatible individuals on liver transplant waiting list.
    8. Desensitization of HLA Incompatible individuals on renal transplant waiting list when used as a single agent.
    9. Ebstein-Barr Virus infection in any condition other than X-linked immunodeficiency
    10. Glomerulosclerosis, Focal Segmental
    11. Graft-Versus-Host Disease, Acute for treatment of active disease
    12. Graft-Versus-Host Disease, Acute, as a preventative agent
    13. Graft-Versus-Host Disease, Acute, for steroid-refractory disease
    14. Graft-Versus-Host Disease, Chronic, as initial, (first line) therapy
    15. Graft-Versus-Host Disease, Chronic, as preventative therapy
    16. Fibrillary Glomerulonephritis· Hemophilia A, treatment to eradicate or suppress autoimmune anti-factor VIII antibodies.
    17. IgA Nephropathy
    18. Multiple Myeloma, for Primary, Recurrent, Relapsed, or Refractory Disease in individuals with absence of CD20+ antigen plasma cells
    19. Multiple Sclerosis, Primary Progressive
    20. Polymyositis
    21. Pemphigus Vulgaris and Pemphigus Foliaceous, when administered concomitantly (or sequentially) with immune globulin.
    22. Primary Biliary Cirrhosis
    23. Red Cell Aplasia or Diamond Blackfan Anemia
    24. Rheumatoid arthritis, when administered concomitantly (or within the same month) with anti-tumor necrosis alpha drugs.
    25. Transplant, Heart, Desensitization of ABO Incompatible individuals on Transplant Waiting List
    26. Transplant, Kidney, Desensitization of HLA Panel Reactive Antibodies developing in kidney transplant recipients to prevent potential rejection.
    27. Transplant, Kidney, as Induction Given Prior to Transplant
    28. Transplant, Liver, Desensitization of ABO Incompatible individuals on Transplant Waiting List
    29. Transplant, Liver, Rejection
    30. Transplant, Lung, Acute Rejection due to Cellular Vascular Rejection
    31. Transplant, Lung, Desensitization of ABO Incompatible individuals on Transplant Waiting List
    32. Urticaria, Chronic
 
Requests for coverage of any of the conditions on the non-Covered list, or for any condition not listed in the policy will require submission of data on effectiveness which will be reviewed to determine if the proposed indication meets member certificate of benefit coverage criteria.
 
For off-label use for non-malignant conditions, rituximab does not meet member certificate of benefit primary coverage criteria if:
 
    1. The use is being studied in phase I, II, or III clinical trials or otherwise under study to determine effectiveness.
    2.  If there is lack of scientific evidence of effectiveness; or
    3.  The effectiveness of the particular use is in conflict or the subject of debate amongst experts.
 
For contracts without primary coverage criteria, any other use of rituximab that is not listed as a covered indication is investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage.
 
Effective October 19, 2022 to October 25, 2022
 
Meets Primary Coverage Criteria Or Is Covered For Contracts Without Primary Coverage Criteria
 
1. RITUXIMAB (e.g., Rituxan®)
The use of Rituximab meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for the following indications:
 
INITIAL APPROVAL STANDARD REVIEW for up to 12 months:
 
FDA APPROVED INDICATIONS
    1. Rheumatoid Arthritis (RA), For those moderate to severely active, in combination with methotrexate for individuals who have had an inadequate response to one or more TNF antagonist therapies.
    2. Granulomatosis with Polyangiitis (GPA) (Wegener’s Granulomatosis) and Microscopic Polyangiitis (MPA) (Anti-Neutrophil Cytoplasmic Antibody Vasculitis) in combination with Glucocorticoids in adult and pediatric individuals 2 years of age and older in combination with glucocorticoids.
    3. Pemphigus Vulgaris (PV) and Pemphigus Foliaceus, unresponsive to conventional therapy (e.g., systemic corticosteroids and immunosuppressive agents) or in those individuals in whom these drugs would not be tolerated.
 
CONTINUATION OF THERAPY for 12 months:
1. Individual meets criteria for initial approval based on indication.
2. Individual  has experienced a positive clinical response to rituximab.
3. Must be dosed in accordance with the FDA label unless otherwise specified.
 
Dosage and administration
 
Must be  dosed in accordance with the FDA label unless otherwise specified.
 
RA
The dose for RA in combination with methotrexate is two-1,000mg IV infusions separated by 2 weeks, (one course) every 24 weeks, or based on clinical evaluation not sooner than every 16 weeks. Methylprednisolone 100 mg IV or equivalent glucocorticoid is recommended 30 min prior to each infusion.
 
GPA and MPA
Induction dose for adult individuals with active GPA and MPA in combination with glucocorticoids is 375 mg/m2 once weekly for 4 weeks. Follow-up dose for those who have achieved control with induction treatment, in combination with glucocorticoids is 500 mg/m2 IV separated by 2 weeks followed by 500 mg/m2 infusion every 6 months thereafter based on clinical evaluation.
 
Induction dose for pediatric individuals in combination with glucocorticoids is 375 mg/m2 once weekly for 4 weeks. Follow-up dosing in combination with glucocorticoids is two 250 mg/m2 infusions separated by two weeks, followed by a 250 mg/m2 IV infusions every 6 months thereafter based on clinical evaluation.
 
PV
The dose for PV is two-1,000 mg IV infusion separated by 2 weeks in combination with a tapering course of glucocorticoids, then a 500 mg infusion at month 12 and every 6 months thereafter or based on clinical evaluation. Dose on relapse is a 1,000 mg infusion with considerations to resume or increase the glucocorticoid dose based on clinical evaluation. Subsequent infusion may be no sooner than 16 weeks after the previous infusion.
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
2. RITUXIMAB-PVVR (e.g., Ruxience), RITUXIMAB-ARRX (e.g., Riabni), and Rituximab-abbs (e.g., Truxima)
Rituximab-pvvr, Rituximab-arrx, and Rituximab-abbs meet member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for the following indication:
 
FDA APPROVED INDICATIONS
    1. Granulomatosis with Polyangiitis (GPA) (Wegener’s Granulomatosis) and Microsopic Polyangiitis (MPA) in adult Individuals  in combination with glucocorticoids.
    2. Rheumatoid Arthritis (RA) in combination with methotrexate in adult individiuals with moderately to severely active RA who have inadequate response to one or more TNF antagoinist therapies.
 
CONTINUATION OF THERAPY for 12 months:
1. Individual meets criteria for initial approval based on indication.
2. Individual has experienced a positive clinical response to rituximab.
3. Must be dosed in accordance with FDA label unless otherwise specified.
 
Dosage and administration
 
Must be dosed in accordance with the FDA label unless otherwise specified.
 
GPA and MPA
Induction dose for adult individuals with active GPA and MPA in combination with glucocorticoids is 375 mg/m2 once weekly for 4 weeks. Follow-up dose for those who have achieved control with induction treatment, in combination with glucocorticoids is 500 mg/m2 IV separated by 2 weeks followed by 500 mg/m2 infusion every 6 months thereafter based on clinical evaluation.
 
RA
The dose for RA in combination with methotrexate is two-1,000 mg intravenous infusions separated by 2 weeks (one course) every 24 weeeks or based on clinical evaluation, but not sooner than every 16 weeks. Methylprednisolone 100 mg intravenous or equivalent.
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
OFF-LABEL INDICATIONS for Rituximab (e.g., Rituxan) and Biosimilars, (e.g., Ruxience; Truxima and Riabini)
The use of Rituximab and biosimilars meets member benefit certificate primary coverage criteria that there be scientific evidence of effectiveness in improving health outcomes for off-label use for treatment of the following conditions:
 
1. Acquired inhibitors in Hemophilia – (Refractory or intolerant of cyclophosphamides and steroid therapy) [Collins 2009, Rossi 2016]
2. Autoimmune Hemolytic Anemia (AIHA) in individuals that have failed to respond to corticosteroid therapy [Shanafelt 2003, Birgens 2013, Michel 2017]
3. Autoimmune encephalitis (AE) when the following criteria are met [Graus, 2016]
· Autoimmune limbic encephalitis - ALL criteria is required:
o Subacute onset (rapid progression of <3 months) of working memory deficits (short-term memory loss), seizures, or psychiatric symptoms suggesting involvement of the limbic system
o Bilateral brain abnormalities on T2-weighted FLAIR MRI highly restricted to the medial temporal lobes
o At least one of the following:
· CSF pleocytosis (>5 white blood cells per mm3)
· EEG with epileptic or slow-wave activity involving the temporal lobes
· Reasonable exclusion of alternative causes.
· For continued therapy, diagnosis is confirmed by detection of a specific autoantibody associated with AE [including but not limited to:
o NMDAR, LGI1, Caspr2, AMPAR, GABA-A or GABA-B receptor, IgLON5, DPPX, GlyR, mGluR1, mGluR2, mGluR5, Neurexin 3-alpha, or dopamine-2 receptor (D2R)]; AND
o individual has had an inadequate response to, is intolerant of, or has a contraindication to first line agent(s) including immunoglobulin therapy or plasma exchange
4. Anti-NMDA receptor encephalitis (Graus, 2016) – ALL criteria below must be met:
· Rapid onset (<3 months) of at least four of the six following major groups of symptoms:
o Abnormal (psychiatric) behavior or cognitive dysfunction
o Speech dysfunction
o Seizures
o Movement disorder, dyskinesias, or rigidity/abnormal postures
o Decreased level of consciousness
o Autonomic dysfunction or central hypoventilation
· At least one of the following laboratory results:
o Abnormal EEG (focal or diffuse slow or disorganized activity, epileptic activity, or extreme delta brush), OR
o CSF with pleocytosis or oligoclonal bands
· Reasonable exclusion of other disorders
· For continuation of therapy,
o IgG anti-GluN1 antibodies [diagnosis of anti-NMDA receptor encephalitis is confirmed by the detection of IgG antibodies to the GluN1 (also known as NR1) subunit of the NMDA receptor in serum or CSF] in the presence of one or more of the six major groups of symptoms, after reasonable exclusion of other disorders, AND
o individual has had an inadequate response to, is intolerant of, or has a contraindication to first line agent(s) including immunoglobulin therapy or plasma exchange
5. Catastrophic Antiphospholipid antibody Syndrome (CAPS) [Rao 2009]
6. Cryoglobulinemia associated with Sjogren’s or SLE [Ramos 2009]
7. Cryoglobulinemic vasculitis associated with Hepatitis C virus infection [Petrarca 2010, KDIGO 2018]
8. Ebstein-Barr Virus infection in individuals with X-linked Immunodeficiency [Milone 2005]
9. Graft vs. Host Disease, Chronic [Cutler 2006]10.
10. Grave’s Ophthalmopathy [El Fassi 2007, Salvi 2007]
11. Immunoglobulin G4 related disease – (Failure or intolerance to steroid therapy.) [Carruthers 2015]
12. ITP, in children with ITP who have significant ongoing bleeding and/or have a need for improved quality of life despite conventional treatment. Also, may be considered as an alternative to splenectomy in children with chronic ITP or as therapy in those who have failed splenectomy. [Oved 2017
13. ITP, in adults at risk of bleeding who have failed one line of therapy such as corticosteroids, IVIg, or splenectomy. [Ghanima 2015]
14. Lupus Erythematosus, Systemic, as second- or third-line therapy [Merrill 2010]
15. Evan’s Syndrome, pediatric and adult, who are unresponsive to, or unable to tolerate corticosteroids [Bader-Meunier 2007, Rucker 2008]
16. Myasthenia Gravis generalized and MuSK positive Myasthenia Gravis. [Narayanaswami 2020, Singh 2019]
17. Membranous Glomerulonephropathy for patients refractory to, or cannot take alkylating agents or calcineurin inhibitors [Müller-Deile 2015, Anjum 2019]17
18. Membranous Nephropathy (primary MN) In individuals  with primary MN, initial therapy is based on an assessment of the individual's risk of progressive disease [DeVriese, 2017]:
· In individuals  with primary MN regardless of phospholipase A2 receptor (PLA2R) – 2 or more of the following are considered very high risk for progressive disease:
o Serum creatinine 1.5 mg/dL [133 micromol/L] considered to be due to active MN
o Progressive decline in kidney function (eg, decrease in estimated glomerular filtration rate [eGFR] 25 percent from baseline over the prior two years) considered to be due to active MN
o Severe, disabling, or life-threatening nephrotic syndrome (defined by the presence of a serum albumin <2.5 g/dL [if measured by bromocresol green methods] or <2.0 g/dL [if measured by bromocresol purple methods] and refractory edema, or a thromboembolic event) For those individuals with primary MN and normal kidney function and do not have severe or life -threatening nephrotic syndrome, will observe for 3- 6 months (Including renin-angiotensin system inhibition). After 3-6 months of observation
 
High risk – Individuals  with 2 or more of the following for progressive disease:
· Decrease in eGFR of 25 percent, not explained by other causes, at any time during the observation period
· Proteinuria >8 g/day at the end of the observation period or persistent nephrotic syndrome (proteinuria >3.5 g/day and serum albumin <3.5 g/dL [if measured by bromocresol green methods] or <3.0 g/dL [if measured by bromocresol purple methods])
· If the individual is anti-PLA2R antibody positive, serial anti-PLA2R antibody titers are high (arbitrarily defined as 150 RU/mL by the ELISA method) and not declining or are increasing to 150 RU/mL
Moderate risk – pts with 2 or more of the following for progressive disease:
· Normal or stable eGFR (ie, <25 percent decrease) over the three-to-six-month period
· Proteinuria <4 g/day at the end of the observation period
· If the patient is anti-PLA2R antibody positive, serial anti-PLA2R antibody titers are low (arbitrarily defined as <50 RU/mL by the ELISA method) or decreasing by 25 percent at three to six months
19. Monoclonal Gammopathy of Unknown Significance with Polyneuropathy (with or without anti-MAG antibodies in individuals refractory to standard therapy [Chaudry 2017, Svahn, 2018]
20. Neuromyelitis Optica for individuals with disease refractory to immunosuppressive drugs and/or plasmapheresis [Nikoo]  
21. Multiple Sclerosis, Relapsing Remitting [Rae-Grant 2018]
22. Pediatric Nephrotic Syndrome –(Refractory or intolerant to steroid therapy and immunosuppressants.) [KDIGO 2012]
23. Rheumatoid arthritis, as a single agent in individuals who have failed DMARDs, and antitumor necrosis alfa drugs, and who have been shown to be intolerant of methotrexate FDA Approval
24. Sjögren’s Syndrome [Dass 2008]
25. Systemic Sclerosis (SSc)
      • Systemic Sclerosis (SSc)-associated Interstitial Lung Disease (ILD) (Goswami, 2021)
          • Previous trial/failure of standard treatment (i.e., Mycophenolate Mofetil (MMF), cyclophosphamide, or tocilizumab therapy).
          • Baseline pulmonary functional parameters [FVC (% predicted)]
      • Systemic Sclerosis (SSc)-Pulmonary Arterial Hypertension (PAH) (Zamanian, 2021)
        • Screening 6-minute Walking Distance (6MWD) of at least 100 meters
26. Thrombotic Thrombocytopenic Purpura, Acquired, due to ADAMTS13 deficiency - (refractory or relapsing to plasma exchange treatment and steroid therapy.) [Joly 2017]
27. Transplant, Heart, Acute Graft Rejection due to Antibody Mediated Disease, in individuals refractory to plasmapheresis/IVIG [Bierl 2006]
28. Transplant, Kidney, Desensitization of ABO Incompatible Patients on Renal Transplant Waiting List, when used in conjunction with IVIG [Echterdiek 2020, Wongsaroj 2015]
29. Transplant, Kidney, Desensitization of Panel Reactive Antibodies on Renal Transplant Waiting List [Vo 2010]Transplant, Kidney, Acute Graft Rejection due to Antibody Mediated Disease, when used as second-line therapy [KIDGO 2009]
30. Transplant, Lung, Acute Humoral Antibody Mediated Disease [Martinu 2010]
31. Transplant, Heart, Desensitization of Panel Reactive Antibodies in Patients on Cardiac Transplant Waiting List [Colvin, 2019]
 
CONTINUATION OF THERAPY for 12 months:
1. Individual meets criteria for initial approval based on indication.
2. Individual has experienced a positive clinical response to rituximab.
3. Must be dosed in accordance with FDA label unless otherwise specified.
 
Please refer to a separate policy on Site of Care or Site of Service Review (policy #2018030) for pharmacologic/biologic medications.
 
Does Not Meet Primary Coverage Criteria Or Is Investigational For Contracts Without Primary Coverage Criteria
 
The use of Rituximab and biosimilars do not meet member benefit Primary Coverage Criteria that there be scientific evidence of effectiveness in improving health outcomes for any other indications including the following conditions:
 
1. Atopic Eczema
2. Autoimmune Neutropenia
3. Autoimmune Retinopathy
4. Chronic Fatigue Syndrome
5. Chronic Inflammatory Demyelinating Polyneuropathy
6. Cryoglobulinemia for any indication except the use described under covered conditions.
7. Desensitization of HLA Incompatible individuals on liver transplant waiting list.
8. Desensitization of HLA Incompatible individuals on renal transplant waiting list when used as a single agent.
9. Ebstein-Barr Virus infection in any condition other than X-linked immunodeficiency
10. Glomerulosclerosis, Focal Segmental
11. Graft-Versus-Host Disease, Acute for treatment of active disease
12. Graft-Versus-Host Disease, Acute, as a preventative agent
13. Graft-Versus-Host Disease, Acute, for steroid-refractory disease
14. Graft-Versus-Host Disease, Chronic, as initial, (first line) therapy
15. Graft-Versus-Host Disease, Chronic, as preventative therapy
16. Fibrillary Glomerulonephritis· Hemophilia A, treatment to eradicate or suppress autoimmune anti-factor VIII antibodies.
17. IgA Nephropathy
18. Multiple Myeloma, for Primary, Recurrent, Relapsed, or Refractory Disease in individuals with absence of CD20+ antigen plasma cells
19. Multiple Sclerosis, Primary Progressive
20. Polymyositis
21. Pemphigus Vulgaris and Pemphigus Foliaceous, when administered concomitantly (or sequentially) with immune globulin.
22. Primary Biliary Cirrhosis
23. Red Cell Aplasia or Diamond Blackfan Anemia
24. Rheumatoid arthritis, when administered concomitantly (or within the same month) with anti-tumor necrosis alpha drugs.
25. Transplant, Heart, Desensitization of ABO Incompatible individuals on Transplant Waiting List
26. Transplant, Kidney, Desensitization of HLA Panel Reactive Antibodies developing in kidney transplant recipients to prevent potential rejection.
27. Transplant, Kidney, as Induction Given Prior to Transplant
28. Transplant, Liver, Desensitization of ABO Incompatible individuals on Transplant Waiting List
29. Transplant, Liver, Rejection
30. Transplant, Lung, Acute Rejection due to Cellular Vascular Rejection
31. Transplant, Lung, Desensitization of ABO Incompatible individuals on Transplant Waiting List
32. Urticaria, Chronic
 
Requests for coverage of any of the conditions on the non-Covered list, or for any condition not listed in the policy will require submission of data on effectiveness which will be reviewed to determine if the proposed indication meets member certificate of benefit coverage criteria.
 
For off-label use for non-malignant conditions, rituximab does not meet member certificate of benefit primary coverage criteria if:
 
      1.  The use is being studied in phase I, II, or III clinical trials or otherwise under study to determine effectiveness.
      2.  If there is lack of scientific evidence of effectiveness; or
      3.  The effectiveness of the particular use is in conflict or the subject of debate amongst experts.
 
For contracts without primary coverage criteria, any other use of rituximab that is not listed as a covered indication is investigational. Investigational services are specific contract exclusions in most member benefit certificates of coverage
 
Due to the detail of the policy statement, the document containing the coverage statements for dates prior to October 19, 2022 are not online. If you would like a hardcopy print, please email: codespecificinquiry@arkbluecross.com .
 
For coverage prior to November 1, 2021, please refer to policy number 2006016.

Rationale:
Due to the detail of the rationale, it is not online. If you would like a hardcopy print, please email : codespecificinquiry@arkbluecross.com
 
Update August 2019
Granulomatosis with polyangiitis (formerly called Wegener’s granulomatosis), microscopic polyangiitis, and renal-limited antineutrophil cytoplasm antibody (ANCA)–associated vasculitides are the main ANCA-associated vasculitis variants.  The combination of cyclophosphamide and glucocorticoids leads to remission in most patients with antineutrophil cytoplasm antibody (ANCA)-associated vasculitides. However, even when patients receive maintenance treatment with azathioprine or methotrexate, the relapse rate remains high. Rituximab may help to maintain remission.
 
In this study, patients with newly diagnosed or relapsing granulomatosis with polyangiitis, microscopic polyangiitis, or renal-limited ANCA-associated vasculitis in complete remission after a cyclophosphamide-glucocorticoid regimen were randomly assigned to receive either 500 mg of rituximab on days 0 and 14 and at months 6, 12, and 18 after study entry or daily azathioprine until month 22. The primary end point at month 28 was the rate of major relapse (the reappearance of disease activity or worsening, with a Birmingham Vasculitis Activity Score>0, and involvement of one or more major organs, disease-related life-threatening events, or both).
 
115 enrolled patients (87 with granulomatosis with polyangiitis, 23 with microscopic polyangiitis, and 5 with renal-limited ANCA-associated vasculitis) received azathioprine (58 patients) or rituximab (57 patients). At month 28, major relapse had occurred in 17 patients in the azathioprine group (29%) and in 3 patients in the rituximab group (5%) (hazard ratio for relapse, 6.61; 95% confidence interval, 1.56 to 27.96; P=0.002). The frequencies of severe adverse events were similar in the two groups. Twenty-five patients in each group (P=0.92) had severe adverse events; there were 44 events in the azathioprine group and 45 in the rituximab group. Eight patients in the azathioprine group and 11 in the rituximab group had severe infections, and cancer developed in 2 patients in the azathioprine group and 1 in the rituximab group. Two patients in the azathioprine group died (1 from sepsis and 1 from pancreatic cancer) (Guillevin L, et al 2014).
In conclusion, the between-group differences in relapse rate observed at month 28 in this trial showed that 500-mg rituximab infusions administered every 6 months were superior to azathioprine as maintenance therapy for ANCA-associated vasculitides, at least for patients positive for anti–proteinase 3 ANCA. Rituximab use for maintenance in those patients was found to have a clear clinical benefit in our study. Further studies are warranted for patients with antimyeloperoxidase ANCA–positive vasculitis. (Funded by the French Ministry of Health; MAINRITSAN ClinicalTrials.gov number, NCT00748644; EudraCT number, 2008-002846-51.).
 
Granulomatosis with polyangiitis (GPA) is a systemic necrotizing vasculitis, typically engaging the upper airways, kidneys and lungs and often associated with circulating anti-neutrophil cytoplasmic antibodies (ANCA) directed against proteinase 3 (PR3). The well-established standard treatment for remission induction in GPA has been cyclophosphamide (CY), in combination with corticosteroids (CS) [Hoffman GS, et al. 1992 and Mukhtyar C, et al. 2009]. This treatment regimen, introduced in the 1970s, has dramatically improved the outcome for GPA patients but with a risk of considerable side effects, including infections, sterility and bladder cancer [Knight A, et al. 2004]. Maintenance treatment is usually given with methotrexate (MTX), azathioprine (AZA) or mycophenolate mofetil (MMF), but as at least 50 % of patients have one or several relapses, repeated induction treatment is often necessary with the risk of high cumulative doses of CY [Jayne D et al 2003 and Pagnoux C, et al. 2008].
 
Recently, rituximab (RTX) has been approved by the FDA and the European Medicines Agency (EMA) for induction treatment of GPA and microscopic polyangiitis (MPA) in combination with CS, using the lymphoma protocol of 375 mg/m2 once weekly for 4 weeks. However, the efficacy and safety of repeated RTX courses as maintenance treatment has not yet been established.
 
The study includes 12 patients (seven females, five males) with relapsing GPA treated with repeated cycles of RTX from January 2003 through February 2013. Of the 12 included patients, all with a positive proteinase 3–anti-neutrophil cytoplasmic antibodies, generalised disease and a median disease duration of 35 months (21–270), 92% (11/12) achieved sustainable remission during a median follow-up time of 32 months (range 21–111) from first RTX treatment. Concomitant immunosuppressants were reduced. Infections were the most common adverse events, but infections were an issue also before the start of RTX. RTX administered every 6 months seems to be an effective maintenance treatment in a population with severe, relapsing long-standing GPA. Granulomatous as well as vasculitic manifestations responded equally well. Infections are a problem in this patient group but no new safety problems were identified [Knight A, et al. 2014].
 
Update October 2019
 The study design consisted of an initial 6-month remission induction phase, and a minimum 12month follow-up phase up to a maximum of 54 months (4.5 years) in pediatric patients 2 years to 17 years of age with GPA and MPA. Patients were to receive a minimum of 3 doses of intravenous methylprednisolone (30 mg/kg/day, not exceeding 1g/day) prior to the first RITUXAN or non-U.S.licensed rituximab intravenous infusion. The remission induction regimen consisted of four once weekly intravenous infusions of RITUXAN or non-U.S.-licensed rituximab at a dose of 375 mg/m2 BSA, on study days 1, 8, 15 and 22 in combination with oral prednisolone or prednisone at 1 mg/kg/day (max 60 mg/day) tapered to 0.2 mg/kg/day minimum (max 10 mg/day) by Month 6. After the remission induction phase, patients could receive subsequent RITUXAN or non-U.S.licensed rituximab intravenous infusions on or after Month 6 to maintain remission and control disease activity. The primary objectives of this study were to evaluate safety and PK parameters in pediatric GPA and MPA patients (2 years to 17 years of age). The efficacy objectives of the study were exploratory and principally assessed using the Pediatric Vasculitis Activity Score (PVAS).
A total of 25 pediatric patients 6 years to 17 years of age with active GPA and MPA were treated with RITUXAN or non-U.S.-licensed rituximab in a multicenter, open-label, single-arm, uncontrolled study (NCT01750697). The median age of patients in the study was 14 years and the majority of patients (20/25 [80%]) were female.
 
All 25 patients completed all four once weekly intravenous infusions for the 6-month remission induction phase. A total of 24 out of 25 patients completed at least 18 months from Day 1 (baseline). After the 6-month remission induction phase, patients who had not achieved remission or who had progressive disease or flare that could not be controlled by glucocorticoids alone received additional treatment for GPA and MPA, that could include RITUXAN or non-U.S.-licensed rituximab and/or other therapies, at the discretion of the investigator. Planned follow-up was until Month 18 (from Day 1).
Fourteen out of 25 patients (56%) received additional RITUXAN or non-U.S.-licensed rituximab treatment at or post Month 6, up to Month 18. Five of these patients received four once weekly doses of intravenous RITUXAN or non-U.S.-licensed rituximab approximately every 6 months; 5 of these patients received a single dose of RITUXAN or non-U.S.licensed rituximab every 6 months, and 4 of these patients received various other RITUXAN or non-U.S.-licensed rituximab doses/regimens according to investigator. Of the 14 patients who received follow-up treatment between Month 6 and Month 18, 4 patients first achieved remission between Months 6 and 12 and 1 patient first achieved remission between Months 12 and 18. Nine of these 14 patients achieved PVAS remission by Month 6 but required additional follow-up treatment after Month 6.
 
December 2019 Update
A study was conducted to retrospectively evaluate the efficacy and safety of rituximab (Rtx) treatment in patients with anti-synthetase syndrome (ASS) and severe interstitial lung disease (ILD).
Patients with severe ILD and>12 months follow-up post-Rtx were identified from the Oslo University Hospital ASS cohort (n = 112). Clinical data, including pulmonary function tests (PFTs), were retrospectively collected from medical reports. Extent of ILD pre-, and post-Rtx was scored on thin-section high-resolution CT (HRCT) images and expressed as a percentage of total lung volume. Muscle strength was evaluated by manual muscle testing of eight muscle groups (MMT8).
 
Altogether, 34/112 ASS patients had received Rtx; 24/34 had severe ILD and>12 months follow-up post-Rtx (median 52 months). In these 24 patients, the median percentage of predicted forced vital capacity, forced expiratory volume in 1 s (FEV1) and diffusing capacity of the lungs for carbon monoxide (DLCO) increased by 24%, 22% and 17%, respectively, post-Rtx. Seven patients (allwith disease duration<12 months and/or acute onset/exacerbation of ILD) had>30% improvement in all three PFTs. HRCT analysis showed a median 34% reduction in ILD extent post-Rtx. MMT8 score increased post-Rtx. During follow-up, 7/34 (21%) Rtx-treated ASS patients died; 6/7 deaths were related to infections. The mortality rate in the Rtx-treated group was comparable to that of the remaining ASS cohort (25/78 deceased; 32%).
 
In conclusion, this study, which included 24 Rtx-treated ASS patients with severe ILD, reports improved PFTs after a median 52 months follow-up post-Rtx. The best outcome was observed in patients with a disease duration<12 months and/or acute onset/exacerbation of ILD. The study indicates that Rtx could be a treatment option for selected ASS patients, but infections should be given attention.
 
A study was conducted to assess the efficacy of rituximab (RTX) in SSc.
Fourteen patients with SSc were evaluated. Eight patients were randomized to receive two cycles of RTX at baseline and 24 weeks [each cycle consisted of four weekly RTX infusions (375 mg/m(2))]in addition to standard treatment, whereas six patients (control group) received standard treatment alone. Lung involvement was assessed by pulmonary function tests (PFTs) and chest high-resolution CT (HRCT). Skin involvement was assessed both clinically and histologically.
 
There was a significant increase of forced vital capacity (FVC) in the RTX group compared with baseline (mean +/- s.d.: 68.13 +/- 19.69 vs 75.63 +/- 19.73, at baseline vs 1-year, respectively, P = 0.0018). The median percentage of improvement of FVC in the RTX group was 10.25%, whereas that of deterioration in the controls was 5.04% (P = 0.002). Similarly, diffusing capacity of carbon monoxide (DL(CO)) increased significantly in the RTX group compared with baseline (mean +/- s.d.: 52.25 +/- 20.71 vs 62 +/- 23.21, at baseline vs 1-year respectively, P = 0.017). The median percentage of improvement of DL(CO) in the RTX group was 19.46%, whereas that of deterioration in the control group was 7.5% (P = 0.023). Skin thickening, assessed with the Modified Rodnan Skin Score (MRSS), improved significantly in the RTX group compared with the baseline score (mean +/- s.d.: 13.5 +/- 6.84 vs 8.37 +/- 6.45 at baseline vs 1-year, respectively, P<0.001).
 
In conclusion, results indicate that RTX may improve lung function in patients with SSc. To confirm the results, a proposal of a larger scale, multi-center study with longer evaluation periods is needed. Patients’ with refractory interstitial lung disease, which had previously failed to respond to prednisone and/or other cytotoxic drugs, experiences using rituximab as therapy for refractory antisynthetase syndrome (ASS)-associated interstitial lung disease were assessed retrospectively.
 
All patients (7) received rituximab therapy. Data on pulmonary symptoms, pulmonary function tests and high resolution computed tomography (HRCT) scan of the lungs were collected: (1) before rituximab initiation; and (2) at 6-month and one-year follow-up after the first infusion of rituximab.
 
At one-year follow-up, ASS patients had resolution or improvement of pulmonary clinical manifestations. Patients also exhibited significant improvement of interstitial lung disease parameters: 1) on pulmonary function tests: FVC (p = 0.03) and DLCO (p = 2×10(-5)); 2) and HRCT-scan of the lungs. Due to clinical resolution/improvement of interstitial lung disease, the median daily dose of oral prednisone could be reduced in these 7 ASS patients at one-year follow-up, compared with baseline.
 
These findings suggest that rituximab may be a helpful therapy for refractory interstitial lung disease in patients with ASS.
 
Patients’ with refractory interstitial lung disease, which had previously failed to respond to prednisone and/or other cytotoxic drugs, experiences using rituximab as therapy for refractory antisynthetase syndrome (ASS)-associated interstitial lung disease were assessed retrospectively.
All patients (7) received rituximab therapy. Data on pulmonary symptoms, pulmonary function tests and high resolution computed tomography (HRCT) scan of the lungs were collected: (1) before rituximab initiation; and (2) at 6-month and one-year follow-up after the first infusion of rituximab.
At one-year follow-up, ASS patients had resolution or improvement of pulmonary clinical manifestations. Patients also exhibited significant improvement of interstitial lung disease parameters: 1) on pulmonary function tests: FVC (p = 0.03) and DLCO (p = 2×10(-5)); 2) and HRCT-scan of the lungs. Due to clinical resolution/improvement of interstitial lung disease, the median daily dose of oral prednisone could be reduced in these 7 ASS patients at one-year follow-up, compared with baseline.
 
These findings suggest that rituximab may be a helpful therapy for refractory interstitial lung disease in patients with ASS.
  
March 2021 Update
B-cell anomalies play a role in the pathogenesis of membranous nephropathy. B-cell depletion with rituximab may therefore be noninferior to treatment with cyclosporine for inducing and maintaining a complete or partial remission of proteinuria in patients with this condition.
 
Patients were randomly assigned who had membranous nephropathy, proteinuria of at least 5 g per 24 hours, and a quantified creatinine clearance of at least 40 ml per minute per 1.73 m2 of body-surface area and had been receiving angiotensin-system blockade for at least 3 months to receive intravenous rituximab (two infusions, 1000 mg each, administered 14 days apart; repeated at 6 months in case of partial response) or oral cyclosporine (starting at a dose of 3.5 mg per kilogram of body weight per day for 12 months). Patients were followed for 24 months. The primary outcome was a composite of complete or partial remission of proteinuria at 24 months. Laboratory variables and safety were also assessed.
 
A total of 130 patients underwent randomization. At 12 months, 39 of 65 patients (60%) in the rituximab group and 34 of 65 (52%) in the cyclosporine group had a complete or partial remission (risk difference, 8 percentage points; 95% confidence interval [CI], -9 to 25; P = 0.004 for noninferiority). At 24 months, 39 patients (60%) in the rituximab group and 13 (20%) in the cyclosporine group had a complete or partial remission (risk difference, 40 percentage points; 95% CI, 25 to 55; P<0.001 for both noninferiority and superiority). Among patients in remission who tested positive for anti-phospholipase A2 receptor (PLA2R) antibodies, the decline in autoantibodies to anti-PLA2R was faster and of greater magnitude and duration in the rituximab group than in the cyclosporine group. Serious adverse events occurred in 11 patients (17%) in the rituximab group and in 20 (31%) in the cyclosporine group (P = 0.06).
 
Rituximab was noninferior to cyclosporine in inducing complete or partial remission of proteinuria at 12 months and was superior in maintaining proteinuria remission up to 24 months. (Fervenza FC, Appel GB, Barbour SJ, et.al., 2019)
 
July 2022 Update
Tanaka Y, Takeuchi T, Miyasaka N, et.al. (2015) evaluated the efficacy and safety of rituximab in Japanese patients with systemic lupus erythematosus (SLE) and lupus nephritis (LN) who are refractory to conventional immunosuppressive therapy.
 
Eligible patients received rituximab at a dose of 1,000 mg at days 1, 15, 169, and 183, and were followed for 53 weeks after the first dose of rituximab. Overall disease activity was assessed monthly using a British Isles Lupus Assessment Group activity index. Patients with LN (Upr/Ucr 1.0 at study entry) were identified and their renal responses were evaluated according to the criteria proposed by the American College of Rheumatology (ACR) and the Lupus Nephritis Assessment with Rituximab (LUNAR) study.
 
A total of 34 patients were enrolled and received at least one dose of rituximab. Decrease in disease activity was achieved in 16 (76.5%) out of 34 patients. In 17 patients with LN, response rates of 58.8% and 52.9% by ACR and LUNAR criteria, respectively, were seen. Successful steroid tapering was achieved in association with disease remission. Rituximab was well tolerated, and most adverse drug reactions were grade 1–2 in severity.
 
Rituximab is effective for treatment of Japanese patients with SLE and LN refractory to conventional therapy.
 
Iaccarino L, Bartoloni E, Carli L., et.al. (2015), evaluated the efficacy and safety of rituximab (RTX) in patients with systemic lupus erythematosus (SLE) refractory to standard therapy in the clinical practice setting.
 
145 SLE patients (ACR criteria) were treated with RTX in 11 Italian Centres: 118 with two infusions (1 g), two weeks apart; 27 with 4 infusions (375 mg/m2), one week apart, followed in 10 cases by two further doses, after 1 and 2 months. Systemic complete response (CR) was defined as European Consensus Lupus Activity Measurement (ECLAM) score 1 and partial response (PR) as 1< ECLAM 3. Renal CR (RCR) and renal PR (RPR) were defined according to EULAR recommendations for management of lupus nephritis.
 
Data from 134 (92.4%) patients were available. The mean±SD follow-up was 27.3±18.5 months. After the first course of RTX, CR or PR were observed in 85.8% and CR in 45.5% of cases; RCR or RPR in 94.1% and RCR in 30.9% of patients after 12-month follow-up. Disease flares occurred in 35.1% and renal flares in 31.2% of patients during observational period. Among patients retreated, CR or PR were observed in 84.4% and CR in 57.8% of cases. Adverse events, infections, and infusion reactions occurred after first RTX course in 23.8%, 16.4%, and 3.8% of patients and after retreatment in 33.3%, 22.2% and 11.1%, respectively. No severe infusion reactions or deaths occurred.
 
Data from Italian multicentre RTX Registry confirmed the efficacy and safety of RTX in SLE patients refractory to standard treatment in clinical practice setting.
 
July 2022 Update
 Systemic sclerosis (SSc)-pulmonary arterial hypertension (PAH) is one of the most prevalent and deadly forms of PAH. B cells may contribute to SSc pathogenesis. The safety and efficacy of B-cell depletion for SSc-PAH was investigated. In an NIH-sponsored, multicenter, double-blinded, randomized, placebo-controlled, proof-of-concept trial, 57 patients with SSc-PAH on stable-dose standard medical therapy received two infusions of 1,000 mg rituximab or placebo administered 2 weeks apart. The primary outcome measure was the change in 6-minute-walk distance (6MWD) at 24 weeks. Secondary endpoints included safety and invasive hemodynamics. A machine learning approach was applied to predict drug responsiveness.  57 subjects from 2010 to 2018 were randomized. In the primary analysis, using data through Week 24, the adjusted mean change in 6MWD at 24 weeks favored the treatment arm but did not reach statistical significance (23.6 ± 11.1 m vs. 0.5 ± 9.7 m; P = 0.12). Although a negative study, when data through Week 48 were also considered, the estimated change in 6MWD at Week 24 was 25.5 ± 8.8 m for rituximab and 0.4 ± 7.4 m for placebo (P = 0.03). Rituximab treatment appeared to be safe and well tolerated. Low levels of RF (rheumatoid factor), IL-12, and IL-17 were sensitive and specific as favorable predictors of a rituximab response as measured by an improved 6MWD (receiver operating characteristic area under the curve, 0.88-0.95). B-cell depletion therapy is a potentially effective and safe adjuvant treatment for SSc-PAH. Clinical trial registered with www.clinicaltrails.gov (NCT01086540). (Zamanian RT, Badesch D, Chung L, et.al., 2021)
 
Experts from the Scleroderma Clinical Trials Consortium and the Canadian Scleroderma Research group (n = 170) were asked whether they agreed with SSc algorithms from 2012. Two consensus rounds refined agreement; 62, 54, and 48 experts (36%, 32%, and 28%, respectively) completed the first, second, and third surveys, respectively.
 
For treatment of scleroderma renal crisis, 81% of experts agreed (first-, second-, and third-line treatments were angiotensin-converting enzyme inhibitors, then adding calcium-channel blockers [CCBs], then adding angiotensin receptor blockers [ARBs], respectively). For pulmonary arterial hypertension (PAH), 81% of experts agreed (for mild PAH, treatments were phosphodiesterase 5 [PDE5] inhibitors, then endothelin receptor antagonists plus PDE5 inhibitors, then prostanoids, respectively; for severe PAH, prostanoids were first-line treatment). For mild Raynaud's phenomenon (RP), 79% of experts agreed (treatments were CCBs, then adding PDE5 inhibitors, then ARBs or switching to another CCB, respectively; after the third line of treatment, mild RP was deemed severe). For severe RP, the first- through fourth-line treatments were CCBs, then adding PDE5 inhibitors or prostanoids, then adding PDE5 inhibitors (if not added as second-line treatment) or prostanoids (if not added as second-line treatment), then switching to another CCB, respectively. For active treatment of digital ulcers, 66% of experts agreed (first- and second-line treatments were CCBs and PDE5 inhibitors, respectively). For interstitial lung disease, 69% of experts agreed (for induction therapy, treatments were mycophenolate mofetil [MMF], intravenous cyclophosphamide [IV CYC], and rituximab, respectively; for maintenance, first-line treatment was MMF). For skin involvement, 71% of experts agreed (for a modified Rodnan skin thickness score [MRSS] of 24, first- and second-line treatments were methotrexate [MTX] and MMF, respectively; for an MRSS of 32, first- through fourth-line treatments were MMF, MTX, IV CYC, and hematopoietic stem cell transplantation, respectively). For inflammatory arthritis, 79% of experts agreed (first- through fourth-line treatments were MTX, low-dose glucocorticoids, hydroxychloroquine, and rituximab or tocilizumab, respectively). Algorithms for cardiac and gastrointestinal involvement had ≥75% agreement.
 
Total agreement for SSc algorithms was considerable. These algorithms may guide treatment. (Fernández-Codina A, Walker KM, Pope JE; Scleroderma Algorithm Group., 2018)
 
PubMed and Embase were searched to identify studies on SSc-ILD treated with RTX, confined to a predefined inclusion and exclusion criteria. A systematic review and meta-analysis were performed on the included studies on changes in forced vital capacity (FVC) and diffusion capacity of carbon monoxide (DLCO) from baseline to 6 and 12 months of follow-up.
A total of 20 studies (2 randomized controlled trials, 6 prospective studies, 5 retrospective studies and 7 conference abstracts) were included (n = 575). RTX improved FVC from baseline by 4.49% (95% CI 0.25, 8.73) at 6 months and by 7.03% (95% CI 4.37, 9.7) at 12 months. Similarly, RTX improved DLCO by 3.47% (95% CI 0.99, 5.96) at 6 months and 4.08% (95% CI 1.51, 6.65) at 12 months. In the two studies comparing RTX with other immunosuppressants, improvement of FVC by 6 months in the RTX group was 1.03% (95% CI 0.11, 1.94) greater than controls. At the 12-month follow-up, RTX treatment was similar to controls in terms of both FVC and DLCO. Patients treated with RTX had a lower chance of developing infections compared with controls [odds ratio 0.256 (95% CI 0.104, 0.626), I2 = 0%, P = 0.47).
 
Treatment with RTX in SSc-ILD was associated with a significant improvement of both FVC and DLCO during the first year of treatment. RTX use was associated with lower infectious adverse events. (Goswami RP, Ray A, Chatterjee M,et.al., 2021)
 
A total of 51 patients with SSc-associated interstitial lung disease were recruited and treated with RTX (n = 33) or conventional treatment (n = 18). Median follow-up was 4 years (range: 1-7). Conventional treatment consisted of azathioprine (n = 2), methotrexate (n = 6), and mycophenolate mofetil (n = 10).
 
Patients in the RTX group showed an increase in FVC at 2 years (mean ± SD of FVC: 80.60 ± 21.21 vs 86.90 ± 20.56 at baseline vs 2 years, respectively, p = 0.041 compared to baseline). In sharp contrast, patients in the control group had no change in FVC during the first 2 years of follow-up. At the 7-year time point the remaining patients in the RTX group (n = 5) had higher FVC compared to baseline (mean ± SD of FVC: 91.60 ± 14.81, p = 0.158 compared to baseline) in contrast to patients in the control group (n = 9) where FVC deteriorated (p < 0.01, compared to baseline). Direct comparison between the 2 groups showed a significant benefit for the RTX group in FVC (p = 0.013). Improvement of skin thickening was found in both the RTX and the standard treatment group; however, direct comparison between groups strongly favored RTX at all-time points. Adverse events were comparable between groups.
 
Data indicate that RTX has a beneficial effect on lung function and skin fibrosis in patients with SSc. (Daoussis D, Melissaropoulos K, Sakellaropoulos G, et.al., 2017)
 
SSc is characterized by fibrotic changes in the skin and lung, and the mainstay of treatment has been CYC. B cell involvement suggests that rituximab (RTX) may also be of therapeutic benefit. The aim of the study was to compare the efficacy and safety of RTX compared with CYC in retarding the progression of interstitial lung disease and skin manifestations of primary SSc.
 
60 patients of dcSSc, age 18–60 years with skin and lung involvement, were randomly assigned to monthly pulses of CYC 500 mg/m2 or RTX 1000 mg × 2 doses at 0, 15 days. Primary outcomes were forced vital capacity (FVC) percent predicted at 6 months. Secondary outcomes were absolute change in litres (FVC-l) at 6 months; modified Rodnan skin scores at 6 months, 6-min walk test, Medsgers score and new onset or worsening of existing pulmonary hypertension by echocardiographic criteria.
 
The FVC [%mean (S.D.)] in the RTX group improved from 61.30 (11.28) to 67.52 (13.59), while in the CYC group it declined from 59.25 (12.96) to 58.06 (11.23) at 6 months (P = 0.003). The change of FVC was 1.51 (0.45) l to 1.65 (0.47) l in the RTX group, compared with 1.42 (0.49) to 1.42 (0.46) l in the CYC group. The mRSS changed from 21.77 (9.86) to 12.10 (10.14) in the RTX group and 23.83 (9.28) to 18.33 (7.69) in the CYC group after 6 months. Serious adverse events were more common in the CYC group.
 
RTX is a safe and effective alternative to CYC in the primary therapy of skin and lung manifestations of scleroderma. (Sircar G, Goswami RP, Sircar D, 2018)
 
October 2022 Update
In a study group comprised 1,008 RA patients from 2 independent randomized placebo-controlled phase III clinical trials (REFLEX [Randomized Evaluation of Long-Term Efficacy of Rituximab in Rheumatoid Arthritis] and SERENE [Study Evaluating Rituximab's Efficacy in Methotrexate Inadequate Responders]). A novel threshold selection method was used to identify baseline candidate biomarkers present in at least 20% of patients that enriched for placebo-corrected American College of Rheumatology 50% improvement (ACR50 response; a high clinical efficacy bar) at week 24 after the first course of rituximab.
 
The presence of IgM rheumatoid factor (IgM-RF), IgG-RF, IgA-RF, and IgG anti-cyclic citrullinated peptide (anti-CCP) antibodies together with an elevated C-reactive protein (CRP) level were associated with enhanced placebo-corrected ACR50 response rates in the REFLEX patients with RA who had an inadequate response to anti-tumor necrosis factor therapies. These findings were independently replicated using samples from patients in SERENE who had an inadequate response to disease-modifying antirheumatic drug treatment. The combination of an elevated baseline CRP level together with an elevated level of any RF isotype and/or IgG anti-CCP antibodies was further associated with an enhanced benefit to rituximab.
 
The presence of any RF isotype and/or IgG anti-CCP autoantibodies together with an elevated CRP level identifies a subgroup of patients with RA in whom the benefit of rituximab treatment may be enhanced. Although the clinical benefit of rituximab was greater in the biomarker-positive population compared with the biomarker-negative population, the clinical benefit of rituximab compared with placebo was also clinically meaningful in the biomarker-negative population. (Lal P, Su Z, Holweg CT, et.al., 2011)
 
A phase III study evaluated the efficacy and safety of rituximab plus methotrexate (MTX) in patients with active rheumatoid arthritis (RA) who had an inadequate response to MTX and who were naïve to prior biological treatment.
 
 Patients with active disease on stable MTX (10-25 mg/week) were randomized to rituximab 2 x 500 mg (n=168), rituximab 2 x 1000 mg (n=172), or placebo (n=172). From week 24, patients not in remission (Disease Activity Score (28 joints) > or =2.6) received a second course of rituximab; patients initially assigned to placebo switched to rituximab 2 x 500 mg. The primary end point was American College of Rheumatology 20 (ACR20) response at week 24. All patients were followed until week 48.
 
At week 24, both doses of rituximab showed statistically superior efficacy (p<0.0001) to placebo (ACR20: 54%, 51% and 23%; rituximab (2 x 500 mg) + MTX, rituximab (2 x 1000 mg) + MTX and placebo + MTX, respectively). Secondary end points were also significantly improved for both rituximab groups compared with placebo. Further improvements in both rituximab arms were observed from week 24 to week 48. Rituximab + MTX was well tolerated, demonstrating comparable safety to placebo + MTX through to week 24, and between rituximab doses through to week 48.
 
Rituximab (at 2 x 500 mg and 2 x 1000 mg) plus MTX significantly improved clinical outcomes at week 24, which were further improved by week 48. No significant differences in either clinical or safety outcomes were apparent between the rituximab doses. (Emery P, Deodhar A, Rigby WF, et.al., 2011)
 
2023 Update
Annual policy review completed with a literature search using the MEDLINE database through October 2023.

CPT/HCPCS:
C9399Unclassified drugs or biologicals
J3490Unclassified drugs
J9311Injection, rituximab 10 mg and hyaluronidase
J9312Injection, rituximab, 10 mg
J9999Not otherwise classified, antineoplastic drugs
Q5115Injection, rituximab abbs, biosimilar, (truxima), 10 mg
Q5119Injection, rituximab pvvr, biosimilar, (ruxience), 10 mg
Q5123Injection, rituximab-arrx, biosimilar, (riabni), 10 mg

References: Andersson H, Sem M, Lund MB, Aaløkken TM, Günther A, Walle-Hansen R, Garen T, Molberg(2015) Long-term experience with rituximab in anti-synthetase syndrome-related interstitial lung disease. Rheumatology (Oxford). 2015 Aug;54(8):1420-8. Epub 2015 Mar 3

Anjum N, Nabi Z, Alam MA. (2019) Rituximab In The Treatment Of Refractory Idiopathic Membranous Nephropathy In Pakistani Population. J Ayub Med Coll Abbottabad. 2019 Apr-Jun;31(2):265-268. PMID: 31094128.

Araki M, Wada K, Mitsui Y, Kubota R, et al.(2016) The Efficacy of Rituximab in High-risk Renal Transplant Recipients. Acta Med Okayama. 2016 Aug;70(4):295-7

Aranda JM, Scornic JC, Normann SJ, et al (2002) Anti-CD20 monoclonal antibody (rituximab) therapy for acute cardiac humoral rejection: a case report. Transplantation, 2002; 73:907-910.

Aries PM, Helmich B, Voswinkel J, et al.(2006) Lack of efficacy of rituximab in Wegener's granulomatosis with refractory granulomatous manifestations. Ann Rheum Dis, 2006; 65:853-8.

Arnold DM, Dentali F, Crowther MA, et al.(2007) Systematic review: efficacy and safety of rituximab for adults with idiopathic thrombocytopenic purpura. Ann Intern Med 2007;146(1):25-33.CrossRefPubMedWeb of Science Google Scholar

Arnold DM, Dentali F, et al.(2007) Efficacy and safety of rituximab for adults with idiopathic thrombocytopenic purpura. Ann Intern Med, 2007; 146:25-33.

Bader-Meunier B (2007) Rituximab therapy for childhood Evans syndrome. Hematologica, 2007; 92:1691-1694.

Bates JS, Engemann AM, Hammond JM.(2009) Clinical utility of rituximab in chronic graft-versus-host disease. Ann Pharmacother, 2009; 43:316-321.

Becker(2004) Rituximab as treatment for refractory kidney transplant rejection. Am J Transplantation, 2004; 4:996-1001

Berentsen S(2007) Primary chronic cold agglutinin disease: an update on pathogenesis, clinical features and therapy. Hematology, 2007; 12:361-370.

Berentsen S, Ulvestad E, Gjertsen BT, et al.(2004) Rituximab for primary chronic cold agglutinin disease: A prospective study of 37 courses of therapy in 27 patients. Blood, 2004; 103:2925-2928

Berney T, Delis S, Kato T, et al. (2002) Successful treatment of posttransplant lymphoproliferative disease with prolonged rituximab treatment in intestinal transplant recipients. Transplantation, 2002; 74:1000-1006.

Bierl c, Miller B, Prak EL, et al. (2006) Antibody-mediated rejection in heart transplant recipients: potential efficacy of B-cell depletion and antibody removal. Clin Transpl, 2006; 489-496

Birgens H, Frederiksen H, Hasselbalch HC, et al. (2013) A phase III randomized trial comparing glucocorticoid monotherapy versus glucocorticoid and rituximab in patients with autoimmune haemolytic anaemia. Br J Haematol. 2013; 163(3):393-399.

Blaes AH, Morrison VA(2010) Post-transplant lymphoproliferative disorders following solid-organ transplantation. Expert Rev Hematol. Feb 2010;3(1):35-44. PMID 21082932

Blaes AH, Peterson BA, Bartlett N, et al. (2005) Rituximab therapy is effective for posttransplant lymphoproliferative disorders after solid organ transplantation: results of a phase II trial. Cancer. Oct 15, 2005;104(8):1661-1667. PMID 16149091

Bomback AS, Derebail VK, McGregor JG, et al(2009) Rituximab therapy for membranous nephropathy: A systematic review. Clin J Am Soc Nephrol, 2009; 4; 734-744

Bomprezzi R, Postevka E, Campagnolo D, Vollmer TL. (2011) A review of cases of neuromyelitis optica. Neurologist, 2011; 17:98-104

Borie R, Debray MP, Laine C, et al. (2009) Rituximab therapy in autoimmune pulmonary alveolar proteinosis. Eur Respir J, 2009; 33:1503-1506

Bosch X. (2010) Rituximab in ANCA vasculitis and lupus: bittersweet results. Nature Reviews/Nephrology, 2010; 6:137-139

Braendstrup P, Bjerrum OW, Nielsen OJ, et al.(2005) Rituximab chimeric anti-CD20 monoclonal antibody treatment for adult refractory idiopathic thrombocytopenic purpura. Am J Hematol, 2005; 78:275-280.

Braun A, Neumann T, Oelzner P, et al. (2008) Cryoglobulinemia type III with severe neuropathy and immune complex glomerulonephritis: remission after plasmapheresis and rituximab. Rheumatology International, 2008; 28:503-506.

Brogan P, et al. (2018) Pediatric Open-Label Clinical Study of Rituximab for the Treatment of Granulomatosis with Polyangiitis (GPA) and Microscopic Polyangiitis (MPA) [abstract]. Arthritis Rheumatol. 2018; 70 (suppl 10).

Bucin D, Johanson S, Malm T, et al. (2006) transplantation across the antibodies against HLA and ABO. Transpl Int, 2006; 19:239-244.

Carruthers MN, Topazian MD, Khosroshahi A, et al. (2015) Rituximab for IgG4-related disease: a prospective, open-label trial. Annals of the Rheumatic Diseases 2015;74:1171-1177.

Chandra PA, Margulis Y, Schiff C. (2008) Rituximab is useful in the treatment of Felty’s syndrome. Am J Ther, 2008; 15:321-322

Chaudhry HM, Mauermann ML, Rajkumar SV. (2017) Monoclonal Gammopathy-Associated Peripheral Neuropathy: Diagnosis and Management. Mayo Clin Proc. 2017 May;92(5):838-850. doi: 10.1016/j.mayocp.2017.02.003. PMID: 28473042; PMCID: PMC5573223.

Choquet S, Leblond V, Herbrecht R, et al. (2006) Efficacy and safety of rituximab in B-cell post-transplantation lymphoproliferative disorders: results of a prospective multicenter phase 2 study. Blood. Apr 15 2006;107(8):3053-3057. PMID 16254143

Cohen JD. (2008) Successful treatment of psoriatic arthritis with rituximab. Ann Rheu Dis, 2008; 67:1647-1648

Collins M, Navaneethan SD, Chung M, et al. (2008) Rituximab treatment of fibrillary glomerulonephritis. Am J Kidney Dis, 2008; 52:1158-1162.

Collins PW, Mathias M, Hanley J, et al.(2009) Rituximab and immune tolerance in severe hemophilia A: a consecutive national cohort. J Thromb Haemot. 2009; 7(5):787-794.

ColvinMM, CookJL, ChangPP, HsuDT, KiernanMS, KobashigawaJA, LindenfeldJ, MasriSC, MillerDV, RodriguezER, TyanDB, ZeeviA(2019) Am Heart Assoc Heart Failure Transplantation Com of the Council Clinical Card; Council on Cardio Disease in the Young; Council on Cardio Stroke Nursing; and Council on Cardio Surg and Anes A Scientific Statement From the American Heart Association. Circulation. 2019 Mar 19;139(12):e553-e578. doi: 10.1161/CIR.0000000000000598. PMID: 30776902.

Cutler C, Miklos D, Haesook TK, et al.(2006) Rituximab for steroid-refractory chronic graft-versus-host disease. Blood; 108:756-762

D’Arena G, Califano C, Annunziata M, et al(2007) Rituximab for warm-type idiopathic autoimmune hemolytic anemia: a r retrospective study of 11 adult patients. Eur J Haematol, 2007; 79:53-58.

Daoussis D, Liossis SN, Tsamandas AC, Kalogeropoulou C, Kazantzi A, Sirinian C, Karampetsou M, Yiannopoulos G, Andonopoulos AP.(2010) Experience with rituximab in scleroderma: results from a 1-year, proof-of-principle study. Rheumatology (Oxford). 2010;49(2):271. Epub 2009 May 15.

Dass S, Bowman SJ, Vital EM, et al.(2008) Reduction of fatigue in Sjogren syndrome with rituximab: results of a randomized, double-blind, placebo-controlled pilot study. Ann Rheum Dis, 2008; 67:1541-1544

Dass S, Vital EM, Emery P. (2007) Development of psoriasis after B cell depletion with rituximab. Arthritis Rheum, 2007; 56:2715-2718.

DeVriese AS, Glassock RJ, Nath KA, et al.(2017) A Proposal for a Serology-Based Approach to Membranous Nephropathy. J Am Soc Nephrol. 2017;28(2):421. Epub 2016 Oct 24.

Diaz LA.(2007) Rituximab and pemphigus - a therapeutic advance. New Engl J Med, 2007; 357:605-607

Dimopoulos MA, Anagnostopoulos Z, et al. (2005) Predictive factors for response to rituximab in Waldenstrom’s macroglobulinemia Clin Lymphoma, 2005; 5:270-2.

Dimopoulos MA, Merlini G, et al(2005) How we treat Waldenstrom’s macroglobulinemia. Hematologica, 2005; 90:117-125.

Dungarwalla M, Marsh C, Tooze A, et al.(2007) Lack of clin efficacy of rituximab in treatment of autoimmune neutropenia and pure red cell aplasia: implications pathophysiology. Ann Hematol,2007; 86:191-197.

Echterdiek F, Latus J, Schwenger V. (2020) Immunosuppression in sensitized recipients Curr Opin Organ Transplant. 2020 Feb;25(1):80-85. doi: 10.1097/MOT.0000000000000721. PMID: 31815787.

Edwards J, Szczepanski L, et al. (2004) Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. NEJM 2004; 350:2572-81.

Egawa H, Termukai S, Haga H, et al. (2008) Present status of ABO-incompatible living donor liver transplantation in Japan. Hepatology, 2008; 47:11-13

El Fassi D, Nielsen CH, Nonnema S, et al. (2007) B lymphocyte depletion with the monoclonal antibody rituximab in Graves’ disease: a controlled pilot study. J Clin Endocrinol Metab, 2007; 92:1769-1772

El Tal KA, Posner MR, et al.(2006) Rituximab: a monoclonal antibody to CD20 used in the treatment of pemphigus vulgaris. J Am Acad Dermatol, 2006; 44:449-459.

Eleftheriou D, Melo M, Marks SD, et al(2009) Biologic therapy in primary systemic vasculitis of the young. Rheumatology (Oxford), 2009; 48:978-986.

Elstrom RL, Andreadis C, Aqui NA, et al.(2006) Treatment of PTLD with rituximab or chemotherapy Am J Transplant. Mar 2006;6(3):569-576. PMID 16468968

Emery P, Deodhar A, Rigby WF, Isaacs JD, Combe B, Racewicz AJ, Latinis K, Abud-Mendoza C, Szczepanski LJ, Roschmann RA, Chen A, Armstrong GK, Douglass W, Tyrrell H.(2010) Efficacy and safety of different doses and retreatment of rituximab: a randomised, placebo-controlled trial in patients who are biological naive with active rheumatoid arthritis and an inadequate response to methotrexate Ann Rheum Dis. 2010 Sep;69(9):1629-35. doi: 10.1136/ard.2009.119933. Epub 2010 May 20. Erratum in: Ann Rheum Dis. 2011 Aug;70(8):1519. PMID: 20488885; PMCID: PMC2938895.

Engelhardt M, Jakob A, Ruter B, et al.(2002) Severe cold hemagglutinin disease (CHD) successfully treated with rituximab. Blood, 2002; 100:1922-1923.

Erikkson P.(2005) Nine patients with anti-neutrophil cytoplasmic antibody-positive vasculitis successfully treated with rituximab. J Intern Med, 2005; 257:540-548.

Erre GL, Pardini S, Faedda R, et al. (2008) Effect of rituximab on clinical and laboratory features of antiphospholipid syndrome: A case report and review of literature. Lupus, 2008; 17:50-55

Evans RS, Takahashi K, Duane RT. (1951) Primary thrombocytopenic purpura and acquired hemolytic anemia. Arch Int Med, 1951; 87: 48-65.

Evens AM, David KA, Helenowski I, et al.(2010) Multicenter analysis of 80 solid organ transplantation recipients with post-transplantation lymphoproliferative disease: outcomes and prognostic factors in the modern era. J Clin Oncol. Feb 20 2010;28(6):1038-1046. PMID 20085936

Fakhouri F, Vernant JP, Veyradier A, et al. (2005) Efficiency of curative and prophylactic treatment with rituximab in ADAMTS13-deficient thrombotic thrombocytopenic purpura: a study of 11 cases. Blood, 2005; 106:1932-1937.

Fernández-Codina A, Walker KM, Pope JE; Scleroderma Algorithm Group.(2018) Treatment Algorithms for Systemic Sclerosis According to Experts. Treatment Algorithms for Systemic Sclerosis According to Experts.

Fervenza FC, Abraham RS, Erickson SB, et al.(2010) Rituximab therapy in idiopathic membranous nephropathy: a 2-year study. Clin J Am Soc Nephrol, 2010; 5:2188-2198

Fervenza FC, Appel GB, Barbour SJ, et.al. (2019) Rituximab or Cyclosporine in the Treatment of Membranous Nephropathy. N Engl J Med. 2019 Jul 4;381(1):36-46. doi: 10.1056/NEJMoa1814427. PMID: 31269364.

Fervenza FC, Cosio FG, Erickson SB, et al.(2008) Rituximab treatment of idiopathic membranous nephropathy. Kidney International, 2008, 73:117-125.

Floeg J, Barbour S, Cattran D, et.al(2018) Management and treatment of glomerular diseases (part1): conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. DOE: https://doi.org/10.1016/j.kint.2018.10.018

Fluge O, Mella O. (2009) Clinical impact of B-cell depletion with the anti-CD20 antibody rituximab in chronic fatigue syndrome: a preliminary case series. BMC Neurol, 2009; 9:28

Ghanima W, Khelif A, Waage A, Michel M, Tjønnfjord GE, Romdhan NB, Kahrs J, Darne B, Holme PA; RITP study group. (2015) Rituximab as second-line treatment for adult immune thrombocytopenia (the RITP trial): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet. 2015 Apr 25;385(9978):1653-61. doi: 10.1016/S0140-6736(14)61495-1. Epub 2015 Feb 5. PMID: 25662413.

Godeau B, Porcher R, Fain O, et al(2008) Rituximab efficacy and safety in adult splenectomy candidates with chronic immune thrombocytopenic purpura: results of a prospective multicenter phase 2 study. Blood, 2008; 112:999-1004

Gonzalez-Barca E, Domingo-Domenech E, Capote FJ, et al(2007) Prospective phase II trial of extended treatment with rituximab in patients with B-cell post-transplant lymphoproliferative disease. Haematologica. Nov 2007;92(11):1489-1494. PMID 18024397

Gorson KC, Natarajan N, Roper AH, Weinstein R.(2007) Rituximab treatment in patients with IVIg-dependent immune polyneuropathy: a prospective pilot trial. Muscle Nerve, 2007; 2007; 35:66-69.

Goswami RP, Ray A, Chatterjee M, Mukherjee A, Sircar G, Ghosh P.(2021) Rituximab in the treatment of systemic sclerosis-related interstitial lung disease: a systematic review and meta-analysis. Rheumatology (Oxford). 2021 Feb 1;60(2):557-567. doi: 10.1093/rheumatology/keaa550. PMID: 33164098.

Gottardo NG, Baker DL, Willis FR(2003) Successful induction and maintenance of long-term remission in a child with chronic relapsing autoimmune hemolytic anemia using rituximab Pediatric Hematol Oncol, 2003; 20:7:557-561.

Grabler DJ, Levy M, Kerr D, et al(2008) Neuromyelitis optica pathogenesis and aquaporin 4. J of Neuroinflammation, 2008; 5:22

Graus F, Titulaer MJ, Balu R, et al. (2016) A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol 2016; 15:391.

Guillevin L, Pagnoux C, Karras A, et al.(2014) Rituximab versus azathioprine for maintenance in ANCA-associated vasculitis. N Engl J Med. 2014;371(19):1771

Harth M.(2006) Rituximab for Rheumatoid Arthritis. Canadian Agency for Drugs & Technologies in Health. Sept 2006, Issue 89.

Hawker K, O’Connor P, Freedman MS, et al(2009) Rituximab in patients with primary progressive multiple sclerosis. Results of a randomized double-blind placebo-controlled multicenter trial. Ann Neurol, 2009; 66:460-461.

Hensel W, Villalobos M, et al(2005) Pentostatin/cyclophosphamide with or without rituximab: an effective regimen for patients with Waldenstrom’s macroglobulinemia/lymphoplasmacytic lymphoma. Clin Lymphoma Myeloma, 2005; 6:131-5.

Higashida J, Wun T, et al.(2005) Safety and efficacy of rituximab in patients with rheumatoid arthritis refractory to disease modifying antirheumatic drugs and anti-tumor necrosis factor-alpha treatment. J Rheumatol 2005; 32:2067-9.

Hoffman GS, Kerr GS, Leavitt RY, et al(1992) Wegener’s granulomatosis: an analysis of 158 patients. Ann Intern Med. 1992;116:488–498. doi: 10.7326/0003-4819-116-6-488.

Horning SJ, Bartlett NL, Breslin S, et al(2007) Results of a prospective phase II trial of limited and extended rituximab treatment in nodjular lymphocyte predominant Hodgkin’s disease (NLPHD). Blood 2007; 10:198a; Abstract 644

Iaccarino L, Bartoloni E, Carli L, et.al.(2015) Efficacy and safety of off-label use of rituximab in refractory lupus: data from the Italian Multicentre Registry. Clin Exp Rheumatol. 2015 Jul-Aug;33(4):449-56. Epub 2015 Jun 8. PMID: 26053285.

Jackson C, Sirohi B, Cunningham D, et al(2010) Lymphocyte-predominant Hodgkin lymphoma – clinical features and treatment outcomes from a 30-year experience. Ann Oncol, 2010, Mar 23. (E-pub ahead of print)

Jacob A, Weinshenker BG, Violich I, et al.(2008) Treatment of neuromyelitis optica with rituximab. Retrospective analysis of 25 patients. Arch Neurol, 2008; 65:1443-1448

Jagadeesh D, Woda BA, Draper J, et al.(2012) Post transplant lymphoproliferative disorders: risk, classification, and therapeutic recommendations. Curr Treat Options Oncol. Mar 2012;13(1):122-136. PMID 22241590

Jayne D, Rasmussen N, Andrassy K, et al.(2003) A randomized trial of maintenance therapy for vasculitis associated with antineutrophil cytoplasmic autoantibodies. N Engl J Med. 2003;349:36–44. doi: 10.1056/NEJMoa020286

Joly BS, Coppo P, Veyradier A. (2017) Thrombotic thrombocytopenic purpura. Blood. 2017 May 25;129(21):2836-2846. doi: 10.1182/blood-2016-10-709857. Epub 2017 Apr 17. PMID: 28416507.

Joly P, Maho-Vaillant M, Prost-Squarcioni C et al.(2017) First-line ritux-imab combined with short-term prednisone versus prednisonealone for the treatment of pemphigus (Ritux 3): a prospective,multicentre, parallel-group, open-label randomised trial. Lancet 2017; 389:2031–40.2

Joly P, Mouquet H, Roujeau J-C.(2007) A single cycle of rituximab for the treatment of severe pemphigus. New Engl J Med, 2007; 357:545-552.

Jones RB, Ferraro AJ, Chaudhry AN, et al.(2009) A multicenter survey of rituximab therapy for refractory antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis & Rheumatism, 2009; 60:2156-2168

Jones RB, Tervaert JWC, Hauser T, et al for the European Vasculitis Study Group(2010) Rituximab versus cyclophosphamide in ANCA-associated renal vasculitis. New Engl J Med, 2010; 361:211-220.

Kaczmarek I, Deutsch MA, Sadoni S, et al.(2007) Successful management of antibody-mediated cardiac allograft rejection with combined immunoadsorption and anti-CD20 monoclonal antibody treatment: case report and literature review. J Heart Lung Transplant, 2007; 26:511-515

Kahl BS, Hong F, Williams ME, et al.(2014) Rituximab extended schedule or re-treatment trial for low-tumor burden follicular lymphoma: eastern cooperative oncology group protocol e4402. J Clin Oncol. Oct 1 2014;32(28):3096-3102. PMID 25154829

Kapoor, PT Greipp, WG Morice, et al.(2008) Anti-CD20 monoclonal antibody therapy in multiple myeloma. British J of Hematology, 2008

Katoh N, Matsuda M, Ishii W, et al(2010) Successful treatment with rituximab in a patient with stiff-person syndrome complicated by dysthyroid ophthalmopathy. Intern Med, 2010; 49:237-241

Kavuru MS, Malur A, Marshall I, et al.(2011) An open-label trial of rituximab therapy in pulmonary alveolar proteinosis. Eur Respir J, 2011, Epub ahead of print, Apr 8 2011

KDIGO (Kidney Disease Improving Global Outcomes) (2021) clinical practice guideline for glomerulonephritis (GN). June 2012 Available at: http://kdigo.org/home/glomerulonephritis-gn/. Accessed on: August 31, 2021.

Keogh KA, Wylam ME, Stone JH, Specks U(2005) Induction of remission by B lymphocyte depletion in eleven patients with refractory antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis & Rheumatism, 2005; 52:262-268.

Keogh KA, Ytterberg SR, Specks U., et al(2006) Rituximab for refractory Wegener’s granulomatosis. Am J Respir Crit Care Med, 2006; 173:180-187

Keren A, Hayes HM, O’Driscoll G(2006) Late humoral rejection in a cardiac transplant recipient treated with the anti-CD20 monoclonal antibody rituximab. Transplant Proc, 2006; 38:1520-1522.

Kharfan-Dabaja MA, Mhaskar AR, Djulbegovic B, et al.(2009) Efficacy of rituximab in the setting of steroid-refractory chronic graft-versus-host disease: a systematic review and meta-analysis. Biol Blood Marrow Transplant, 2009; 15:1005-1113

Kidney Disease: (2009) Improving Global Outcomes (KDIGO) Transplant Work Group: KDIGO clinical practice guideline for the care of kidney transplant recipients. Am J Transplant 9[Suppl 3]: S1–S155, 2009

Knight A, Askling J, Granath F, Sparen P, Ekbom A(2004) Urinary bladder cancer in Wegener's granulomatosis: risks and relation to cyclophosphamide. Ann Rheum Dis. 2004;63:1307–1311. doi: 10.1136/ard.2003.019125.

Knight A, Hallenberg H, Baecklund E.(2014) Efficacy and safety of rituximab as maintenance therapy for relapsing granulomatosis with polyangiitis-a case series. Clin Rheumatol, 2014; 33(6): 841-848. Pub online 2013 Aug 20. doi: 10.1007/s10067-013-2351-y.

Kotani T, Takeuchi T, Kawasaki Y, et al.(2006) Successful treatment of cold agglutinin disease with anti-CD20 antibody (rituximab) in a patient with systemic lupus erythematosus. Lupus, 2006; 15:683-685.

Lal P, Su Z, Holweg CT, Silverman GJ, Schwartzman S, Kelman A, Read S, Spaniolo G, Monroe JG, Behrens TW, Townsend MJ.(2011) Inflammation and autoantibody markers identify rheumatoid arthritis patients with enhanced clinical benefit following rituximab treatment. Arthritis Rheum. 2011 Dec;63(12):3681-91. doi: 10.1002/art.30596. PMID: 22127691.

Lamprecht P, Lerin-Lozano C, Merz H, et al.(2003) Rituximab induces remission in refractory HCV associated cryoglobulinemic vasculitis. Ann Rheum Dis, 2003; 62:1230-1233

Landon-Cardinal O, Friedman D, Guiguet M, et al(2018) Efficacy of Rituximab in Refractory Generalized anti-AChR Myasthenia Gravis J Neuromuscul Dis 2018;5(2):241-249.

Lee A, LaCasce AS(2009) Nodular lymphocyte predominant Hodgkin lymphoma. Oncologist, 2009; 14:739-751

Lee PC, Terasaki PI, Takemoto SK, et al.(2002) All chronic reaction failures in kidney transplants were preceded by the development of HLA antibodies. Transplantation, 2002; 74:1192-1194

Leen WG, Weemaes CM, Verbeek MM, et al.(2008) Rituximab and intravenous immunoglobulins for relapsing postinfectious opsoclonus-myoclonus syndrome. Pediatric Neurol, 2008; 39:213-217

Lekharaju V, Chattopadhyay C.(2008) Efficacy of rituximab in Felty’s syndrome. Ann Rheum Dis, 2008; 67:1352

Levine TD(2005) Rituximab in the treatment of dermatomyositis: an open-label pilot study. Arthritis Rheum, 2005; 52:601-607

Lindberg C, Bokarewa, M(2010) Rituximab for severe myasthenia gravis – experience from five patients Acta Neurol Scand, 2010 (Epub ahead of print)

Lovric S, Erdbruegger U, Kumpers P, et al(2009) Rituximab as rescue therapy in anti-neutrophil cytoplasmic antibody-associated vasculitis: a single-centre experience with 15 patients. Nephrol Dial Transplant, 2009; 23:179-185

Lu TYT, Ng KP, Cambridge G, et al.(2009) A retrospective seven-year analysis of the use of B cell depletion therapy in systematic lupus erythematosus at University College London Hospital: the first fifty patients. Arthritis Rheum, 2009; 61:482-487

Lucey MR, Terrault N, Ojo L, et al.(2013) Long-term management of the successful adult liver transplant: 2012 practice guideline by the American Association for the Study of Liver Diseases and the American Society of Transplantation. Liver Transpl. Jan 2013;19(1):3-26. PMID 23281277

Malloy ES, Cabrese LH.(2008) Progressive multifocal leukoencephalopathy in patients with rheumatic diseases: are patients with systemic lupus erythematosus at particular risk Autoimmun Rev, Aug 2008

Mansi, IA, Opran A, and Rosner F.(2002) ANCA-Associated Small-Vessel Vasculitis. Am Fam Physicia. 2002;65(8):1615-1621

Marie I, Dominique S, Janvresse A, Levesque H, Menard JF(2012) Rituximab therapy for refractory interstitial lung disease related to antisynthetase syndrome. Respir Med. 2012 Apr;106(4):581-7. Epub 2012 Jan 24.

Markatseli TE, Kaltsonoudis ES, Voulgari PV, et al(2009) Induction of psoriatic skin lesions in a patient with rheumatoid arthritis treated with rituximab. Clin Exp Rheumatol, 2009; 27:996-998

Martinez-Calle N, Alfonso A, Rifon J, et al(2017) First-line use of rituximab correlates with increased overall survival in late post-transplant lymphoproliferative disorders: retrospective, single-centre study. Eur J Haematol. 2017 Jan;98(1):38-43. doi: 10.1111/ejh.12782. Epub 2016 Jun 28.

Martinu T, Chen, D-F, Palmer SM(2010) Acute cellular rejection and humoral sensitization in lung transplant recipients. Semin Respir Crit Care Med, 2010; 31:179-188,

Maury S, Huguet F, Legusy T, et al(2010) Adverse prognostic significance of CD20 expression in adults with Philadelphia chromosome-negative B-cell precursor acute lymphoblastic leukemia. Hematologica, 2010; 95:324-328

Medeot M, Zaja F, Vianelli N, et al(2008) Rituximab therapy in adult patients with relapsed or refractory immune thrombocytopenic purpura: long-term follow-up results. Eur J Haematol 2008;81

Meijer JM, Pijpe J, Vissink A, et al.(2009) Treatment of primary Sjogren syndrome with rituximab: extended follow-up, safety and efficacy of retreatment. Ann Rheum Dis, 2009; 68:284-285

Merrill JT, Neuwell CM, Wallace DJ, et al.(2010) Efficacy and safety of rituximab in moderately-to-severely active systemic lupus erythematosus: the randomized, double-blind, phase II/III systemic lupus erythematosus evaluation of rituximab trial (EXPLORER). Arthritis Rheum, 2010; 62:222-233

Michel M, Chanet V, Dechartres A, et al.(2009) The spectrum of Evan’s syndrome in adults: new insight into the disease based on the analysis of 68 cases. Blood, 2009

Michel M, Terriou L, Roudot-Thoraval F, et al. (2017) A randomized and double-blind controlled trial evaluating the safety and efficacy of rituximab for warm auto-immune hemolytic anemia in adults (the RAIHA study). Am J Hematol. 2017; 92(1):23-27

Milone MC, Tsai DE, Hodinka RL, et al. (2005) Treatment of primary Epstein-Barr virus infection in patients with X-linked lymphoproliferative disease using B-cell-directed therapy. Blood. 2005;105(3):994. 

Mukhtyar C, Guillevin L, Cid MC, Dasgupta B, de Groot K, Gross W, et al. for the European Vasculitis Study Group(2009) EULAR recommendations for the management of primary small and medium vessel vasculitis. Ann Rheum Dis. 2009;68:310–317. doi: 10.1136/ard.2008.088096.

Müller-Deile J, Schiffer L, Hiss M, Haller H, Schiffer M. (2015) A new rescue regimen with plasma exchange and rituximab in high-risk membranous glomerulonephritis. Eur J Clin Invest. 2015 Dec;45(12):1260-9. doi: 10.1111/eci.12545. Epub 2015 Nov 9. PMID: 26444294.

Mulley WR, Hudson FJ, Tait BD, et al(2009) A single low-fixed dose of rituximab to salvage renal transplants from refractory antibody-mediated rejection. Transplantation, 2009; 87:286-289

Narayanaswami P, Sanders DB, Wolfe G, et al. (2020) International Consensus Guidance for Management of Myasthenia Gravis: 2020 Update. Neurology. 96(3):114-122. doi:10.1212/WNL.0000000000011124

Nasr SH, Valeri AM, Cornell LD, et al.(2011) Fibrillary glomerulonephritis: a report of 66 cases from a single institution. Clin J am Soc Nephrol, 2011; 6:775-784.

Nassi L, Gaidano G. II(2015) Challenges in the management of post-transplant lymphoproliferative disorder. Hematol Oncol. Jun 2015;33 Suppl 1:96-99. PMID 26062065

Nelson RP, Pascuzzi RM, Kessler K, et al(2009) Rituximab for the treatment of thymoma-associated and de novo myasthenia gravis: 3 cases and review. J Clin Neuromuscul Dis, 2009; 10:170-177

Neunert C, Lim W, Crowther M, et al(2011) The American Society of Hematology 2011 evidence-based practice guideline for immune thrombocytopenia Blood 2011 117:4190-4207; doi: https://doi.org/10.1182/blood-2010-08-302984

Nikoo Z, Badihian S, Shaygannejad V, Asgari N, Ashtari F. (2017) Comparison of the efficacy of azathioprine and rituximab in neuromyelitis optica spectrum disorder: a randomized clinical trial J Neurol. 2017 Sep;264(9):2003-2009. doi: 10.1007/s00415-017-8590-0. Epub 2017 Aug 22. PMID: 28831548.

Noss EH, Hausner-Sypek DL, Weinblatt ME.(2006) Rituximab as therapy for refractory polymyositis and dermatomyositis J Rheumatol, 2006; 33:1021-1026

Oertel S, Anagnostopoulos I, Bechstein W, et al(2000) Treatment of posttransplant lymphoproliferative disorder with the anti-CD20 monoclonal antibody rituximab alone in an adult after liver transplantation. Transplantation, 2000; 69:430-432

Oertel SH, Verschuuren E, Reinke P, et al.(2005) Effect of anti-CD 20 antibody rituximab in patients with posttransplant lymphoproliferative disorder (PTLD) Am J Transplant. Dec 2005;5(12):2901-2906. PMID 16303003

Oved JH, Lee CSY, Bussel JB. (2017) Treatment of Children with Persistent and Chronic Idiopathic Thrombocytopenic Purpura: 4 Infusions of Rituximab and Three 4-Day Cycles of Dexamethasone. J Pediatr. 2017 Dec;191:225-231. doi: 10.1016/j.jpeds.2017.08.036. PMID: 29173312; PMCID: PMC6020036.

Pagnoux C, Hogan SL, Chin H, et al(2008) Predictors of treatment resistance and relapse in anti neutrophil cytoplasmic antibody-associated small-vessel vasculitis: comparison of two independent cohorts. Arthritis Rheum. 2008;58:2908–2918. doi: 10.1002/art.23800.

Patel V, Mihatov N, Cooper N, et al.(2007) Long-term follow-up of patients with immune thrombocytopenic purpura whose initial response to rituximab lasted a minimum of one year. J Support Oncol 2007;5 4 suppl 2:82-84. 2007.Google Scholar

Paya CV, Fung JJ, Nalesnik MA, et al.(1999) Epstein-Barr virus and induced post-transplant lymphoproliferative disorders, ASTS/ASTP EBV-PTLD Task Force and The Mayo Clinic Organized International Consensus Development Meeting. Transplantation, 1999; 68:1517-1525

Pestronk A, Florence J, Miller T, et al(2003) Treatment of IgM antibody associated polyneuropathies. J Neurol Neurosurg Psychiatry, 2003; 74:485-489

Petrarca A, Rigacci L, Caini P, et al.(2010) Safety and efficacy of rituximab in patients with hepatitis C virus related mixed cryoglobulinemia and severe liver disease. Blood, 2010; Mar 22

Pham P, Wilkinson A, Gritsch P, et al(2002) Monotherapy with the anti-CD20 monoclonal antibody rituximab in a kidney transplant recipient with posttransplant lymphoproliferative disease. Transplant Proc, 2002; 34:1178-1181

Pijpe J, Meijer JM, Bootsma H, et al(2009) Clinical and histologic evidence of salivary gland restoration supports the efficacy of rituximab treatment in Sjögren’s syndrome. Arthritis Rheum, 2009; 60:3251-3256

Pijpe J, van Imhoff GW, Spijkervet FKL, et al.(2005) Rituximab treatment in patients with primary Sjögren’s Syndrome. Arthritis Rheum, 2005; 52:2740-27-50

Pransatelli MR, Tate ED, Travelstead AL, et al(2010) Long-term cerebrospinal fluid and blood lymphocyte dynamics after rituximab for pediatric opsoclonus-myoclonus. J Clin Immunol, 2010; 30:106-113

Pransatelli MR, Travelstead AL, Tate ED, et al.(2004) B-cell expansion in opsoclonus-myoclonus: effect of rituximab, a biomarker of disease activity. Mov Disorder, 2004; 19:770-777

Pranzatelli MR, Tate ED, Travelstead AL, et al(2005) Immunologic and clinical responses to rituximab in a child with opsoclonus-myoclonus syndrome. Pediatrics, 2005; 115:e115-e119

Pranzatelli MR, Tate ED, Travelstead AL, et al.(2006) Rituximab (anti-CD20) adjunctive therapy for opsoclonus-myoclonus syndrome. J Pediat Hematol Oncol, 2006; 28:585-593

Prayson RA, Frater JL(2002) Rasmussen encephalitis: a clinicopathologic and immunohistochemical study of seven patients. Am J Clin Pathol, 2002; 117:776-782

Provan D, Butler T, Evangelita L, et al(2007) Activity and safety profile of low-dose rituximab for the treatment of autoimmune cytopenias in adults. Hematologica,2007; 92:1695-1698

Quartuccio L, Soardo G, Romano G, et al.(2006) Rituximab treatment for glomerulonephritis in HCV-associated mixed cryoglobulinemia. Rheumatology (Oxford), 2006; 45:842-846

Rae-Grant A, Day GS, Marrie RA, Rabinstein A, et al. (2018) Practice guideline recommendations summary: Disease-modifying therapies for adults with multiple sclerosis: Subcommittee of the American Academy of Neurology. Neurology. 2018 Apr 24;90(17):777-788. doi: 10.1212/WNL.0000000000005347. Erratum in: Neurology. 2019 Jan 8;92(2):112. PMID: 29686116.

Ramanathan S, Koutts J, Hertzberg MS(2005) Two cases of refractory warm autoimmune hemolytic anemia treated with rituximab Am J Hematol, 2005;78:123-126

Ramchandren S(2009) Monoclonal gammopathy and neuropathy Curr Opin Neurol, 2009; 22:480-485

Ramos-Casals M, Soto MJ, Cuadrado MJ, Khamashta MA. (2009) Rituximab in systemic lupus erythematosus: a systematic review of offlabel use in 188 cases. Lupus. 2009; 18(9):767-776

Rao AAN, Arteaga GM, Reed AM, et al(2009) Rituximab for successful management of probable pediatric catastrophic antiphospholipid syndrome. Pediatric Blood Cancer, 2009; 52:536-538

Reams DB, McAdams HP, Howell DN, et al(2003) Posttransplant lymphoproliferative disorder: Incidence, presentation, and response to treatment in lung transplant recipients Chest, 2003; 124:1242-1249

Remuzzi G, Chiurchiu C, et al(2002) Rituximab for idiopathic membranous nephropathy Lancet, 2002; 360:923-5

Rheumatology (Oxford). 2021 Feb 1;60(2):557-567. doi: 10.1093/rheumatology/keaa550. PMID: 33164098.(2016) A multicenter, open-label, comparative study of B-cell depletion therapy with Rituximab for systemic sclerosis-associated interstitial lung disease. Semin Arthritis Rheum. 2017 Apr;46(5):625-631. doi: 10.1016/j.semarthrit.2016.10.003. Epub 2016 Oct 13. PMID: 27839742.

Rios FR, Callejas RJL, Sanches CD, et al.(2009) Rituximab in the treatment of dermatomyositis and other inflammatory myopathies. A report of 49 cases and review of the literature. Clin Exp Rheumatol, 2009; 27:1009-1016

Rituximab for Rheumatoid Arthritis. (2006) Canadian Agency for Drugs & Technologies in Health Sept 2006, Issue 89

Rocatello D(2004) Long-term effects of anti-CD20 monoclonal antibody treatment of cryoglobulinemic glomerulonephritis. Nephrology, Dialysis, Transplantation, 2004; 19:30543061

Rossi B, Blanche P, Roussel-Robert V, et al(2016) Rituximab as first-line therapy for acquired haemophilia A: a single-centre 10-year experience. Haemophilia. 2016; 22(4):e338-41.

Rucker A, Glimm H, Lubbert M, Grullich C.(2008) Successful treatment of life-threatening Evans syndrome due to antiphospholipid antibody syndrome by rituximab-based regimen: a case with long-term follow-up. Lupus, 2008; 17:757-760

Ruegg SJ, Fuhr P, Steck AJ(2004) Rituximab stabilizes multifocal motor neuropathy increasingly less responsive to IVIG Neurology, 2004; 63:2178-2179

Ruggenenti P, Chiurchiu C, Brusegan V, et al(2003) Rituximab in idiopathic membranous nephropathy: a one-year prospective study. J Am Soc Nephrology, 2003; 14:1851-1857

Saadoun D(2008) Rituximab combined with Peg-interferon-ribavirin in refractory hepatitis C virus-associated cryoglobulinemia vasculitis Ann Rheum Dis, 2008; 67:1431-1436

Sailler L.(2008) Rituximab off label use for difficult-to-treat auto-immune diseases: reappraisal of benefits and risks. Clin Rev Allergy Immunol, 2008; 34:103-110

Salvi M, Vannucchi G, Campi I, et al(2007) Treatment of Graves’ disease and associated ophthalmopathy with anti CD20 monoclonal antibody rituximab: an open study European of Endocrinol, 2007; 156-33-40

Sansonno D, De Re V, Lauletta G, et al(2003) Monoclonal antibody treatment of mixed cryoglobulinemia resistant to interferon alpha with an anti-CD20. Blood, 2003; 101:3818-3826

Schollkopf C, Kjeldsen L, Bjerrum OW, et al(2006) Rituximab in chronic cold agglutinin disease: a prospective study of 20 patients. Leuk Lymphoma, 2006; 47:253-260.

Seja F, Baccarani M, Massa P, et al(2010) Dexamethasone plus rituximab yields higher sustained response rates than dexamethasone monotherapy in adults with primary immune thrombocytopenia Blood, 2010; Epub ahead of print

Seo P, Specks U, Keogh KA(2008) Efficacy of rituximab in limited Wegener’s Granulomatosis with refractory granulomatous manifestations J Rheumatol, Aug 2008.

Shaikh A, Habermann TM, Fidler ME, et al(2008) Acurte renal failure secondary to severe type I cryoglobulinemia following rituximab therapy for Waldenstrom’s macroglobulinemia Clin Exp Nephrol, 2008; 12:292-295

Shanafelt TD, Madueme HL, Wolf RC, et al.(2003) Rituximab for immune cytopenia in adults: Idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, and Evans syndrome. Mayo Clinic Proceedings, 2003; 78:1340-1346

Shanafelt TD, Wang XV, Kay NE, et al(2019) Ibrutinib-rituximab or chemoimmunotherapy for chronic lymphocytic leukemia N Engl J Med. 2019 Aug 1;381(5):432-443. PMID 31365801.

Shapiro R(2008) Reducing antibody levels inpatients undergoing transplantation. New Engl J Med, 2008; 359:305-306

Silverman GJ, Weisman S(2003) Rituximab therapy and autoimmune disorders: prospects for anti-B cell therapy. Arthritis Rheum, 2003; 48:184-1492

Silz N, Olson L, McGregor C(2001) Treatment of post-transplant lymphoproliferative disorder with monoclonal CD20 antibody (rituximab) after heart transplantation J Heart Lung Transplant, 2001; 20:770-772.

Simon D, Hosli S, Kostylina F, et al(2008) Anti-CD20 (rituximab) treatment improves atopic eczema J Allergy Clin Immunol,2008; 121:1515-1516

Singavi AK, Harrington AM, Fenske TS.(2015) Post-transplant lymphoproliferative disorders. Singavi AK, Harrington AM, Fenske TS.(2015) Post-transplant lymphoproliferative disorders. Cancer Treat Res. 2015;165:305-27. doi: 10.1007/978-3-319-13150-4_13. PMID: 25655616.

Singh N, Goyal V.(20019) Rituximab as induction therapy in refractory myasthenia gravis: 18 month follow-up study. J Neurol. 2019 Jul; 266(7):1596-1600. doi: 10.1007/s00415-019-09296-y. Epub 2019 Mar 27.

Sircar G, Goswami RP, Sircar D, Ghosh A, Ghosh P,(2018) Intravenous cyclophosphamide vs rituximab for the treatment of early diffuse scleroderma lung disease: open label, randomized, controlled trial, Rheumatology, Volume 57, Issue 12, December 2018, Pages 2106–2113, https://doi.org/10.1093/rheumatology/key213

SL Hauser, Waubant E, Arnold, DL, et al. for the HERMES Trial Group.(2008) B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. New Engl J Med, 2008; 358:676-688.

Sordet C, gottenberg J-E, Hellmich B, et al.(2005) Lack of efficacy of rituximab in Felty’s syndrome. Ann Rheum Dis, 2005; 64:332-333

Stasi R, Pagano A, Stipa E, Amadori S.(2001) Rituximab chimeric anti-CD20 monoclonal antibody treatment for adults with chronic idiopathic thrombocytopenic purpura. Blood, 2001; 98:952-957

Stasi R, Stipa E, Del Poeta G, et al.(2006) Long-term observation of patients with anti-neutrophil cytoplasmic antibody-associated vasculitis treated with rituximab. Rheumatology (Oxford), 2006; 45:1432-1436

Stieglbauer K, Topakian R, Hinterberger G, Aichner FT(2009) Beneficial effect of rituximab monotherapy in multifocal motor neuropathy Neuromuscu Disord, 2009; 19:473-475

Stone JH, Merkel PA, Spiera R, et al, for the RAVE-ITN Research Group.(2010) Rituximab versus cyclophosphamide for ANCA-associated vasculits. New Engl J Med, 2010; 363:221-232.

Summers KM, Kockler DR(2005) Rituximab treatment of refractory rheumatoid arthritis. Ann Pharmaother 2005; 39:2091-5.

Suzuki K, Nagasawa H, Kameda H, et al.(2009) Severe acute thrombotic exacerbation in two cases with anti-phospholipid syndrome after retreatment with rituximab in phase I/II clinical trial for refractory systemic lupus erythematosus. Rheumatology, 2009; 48:198-199

Svahn J, Petiot P, Antoine (2018) J The Francophone anti-MAG cohort Group, et al Anti-MAG antibodies in 202 patients: clinicopathological and therapeutic features Journal of Neurology, Neurosurgery & Psychiatry 2018;89:499-505.

Tahara M, Oeda T, Okada K, Kiriyama T, et al. (2020) Safety and efficacy of rituximab in neuromyelitis optica spectrum disorders (RIN-1 study): a multicentre, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2020 Apr;19(4):298-306. doi: 10.1016/S1474-4422(20)30066-1. Epub 2020 Mar 18. PMID: 32199095.

Takemota SK, Zeevi A, Feng S, et al.(2004) National conference to assess antibody-mediated rejection in solid organ transplantation. Am J Transplantation, 2004; 4:1033-1041

Tanabe K, Ishida H, Shimizu T, et al.(2009) Evaluation of two different preconditioning regimens for ABO-in compatible living kidney donor transplantation. A comparison of splenectomy vs. rituximab-treated non-splenectomy preconditioning regimens. Contributions to Nephrology, 2009; 162:61-74

Tanaka Y, Takeuchi T, Miyasaka N, Sumida T, Mimori T, Koike T, Endo K, Mashino N, Yamamoto K.(2015) Efficacy and safety of rituximab in Japanese patients with systemic lupus erythematosus and lupus nephritis who are refractory to conventional therapy. Mod Rheumatol. 2016;26(1):80-6. doi: 10.3109/14397595.2015.1060665. Epub 2015 Aug 19. PMID: 26054418; PMCID: PMC4732415.

Tandan R, Hehir Mk, Waheed W, et al(2017) Rituximab treatment of myasthenia gravis: A systemic review. Muscle Nerve. 2017 Aug;56(2):185-196.

Teshima T, Nagafuji K, Henzan H, et al.(2009) Rituximab for the treatment of corticosteroid-refractory chronic graft-versus-host disease. Int J Hematol, 2009; 90:253-260.

Thomas DA, O’Brien S, Jorgensen JL, et al.(2009) Prognostic significance of CD20 expression in adults with de novo precursor B-lineage acute lymphoblastic leukemia. Blood, 2009; 113:6330-6337.

Topakian R, Zimprich F, Iglseder S, et al.(2019) High efficacy of rituximab for myasthenia gravis: a comprehensive nationwide study in Austria. J Neurol. 2019 Mar; 266(3):699-706. doi: 10.1007/s00415-019-09191-6. Epub 2019 Jan 16

Tracy JA, Dyck PJ(2010) Investigations and treatment of chronic inflammatory demyelinating polyradiculoneuropathy and other inflammatory demyelinating polyneuropathies. Curr Opin Neurol, 2010 Apr 12 (Epub ahead of print]

Trappe R, Oertel S, Leblond V, et al.(2012) Sequential treatment with rituximab followed by CHOP chemotherapy in adult B-cell post-transplant lymphoproliferative disorder (PTLD): the prospective international multicentre phase 2 PTLD-1 trial. Lancet Oncol. Feb 2012;13(2):196-206. PMID 22173060

Treon SP, Gertz MA, et al.(2006) Update on treatment recommendations from the Third International Workshop on Waldenstrom’s Macroglobulinemia. Blood, 2006;107:3442-3446

Treon SP, Pilarkski LM, Belch AR, et al.(2002) CD20-directed serotherapy in patients with multiple myeloma: biologic considerations and therapeutic applications. J of Immunotherapy, 2002; 25:72-81

Tsokos GC.(2004) B cells, be gone – B-cell depletion in the treatment of rheumatoid arthritis. NEJM 2004; 350:2546-48

Tyden G, Donauer J, Wadstrom J, et al.(2007) Implementation of a protocol for ABO-incompatible kidney transplantation – a three-center experience with 60 consecutive transplantations. Transplantation, 2007; 83:1153-1155

Tyden G, Genberg H, Tollemar , et al(2009) A randomized, double-blind, placebo-controlled, study of single-dose rituximab as induction in renal transplantation. Transplantation, 2009; 15:1325-1329

Usuda M, Fuimori K, Koyamada N, et al.(2005) Successful use of anti-CD20 monoclonal antibody (rituximab) for ABO-incompatible living-related liver transplantation. Transplantation, 2005; 15:12-16

Van Schalk(2006) European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of multifocal motor neuropathy. Eur J Neurol, 2006; 13:802-808

Venhoff N, Rizzi M, Salzer U, et al.(2009) Monozygotic twins with stiff person syndrome and autoimmune thyroiditis: rituximab inefficacy in a double-blind, randomized, placebo controlled crossover study. Ann Rheum Dis, 2009; 68:1506-1508

Vierira CA, Agarwal A, Book BK, et al.(2004) Rituximab for reduction of anti-HLA antibodies in patients awaiting transplantation: Safety, pharmacodynamics, and pharmacokinetics. Transplantation, 2004; 77:542-548

Vo AA, Lukovsky M, Toyoda M, et al.(2008) Rituximab & immune globulin for desensitization during renal transplantation. New Engl J Med, 2008; 359:242-251

Vo AA, Pen G, Toyoda M, et al(2010) Use of intravenous immune globulin and rituximab for desensitization of highly HLA-sensitized patients awaiting kidney transplantation. Transplantation, 2010, Jan 27 [Epub ahead of print].

Weinreb N(2006) Beneficial response to rituximab in refractory Felty’s syndrome. J Clin Rheumatology, 2006; 12:48

Weleber RG(2005) Clinical and electrophysiologic characterization of paraneoplastic and autoimmune retinopathies associated with antienolase antibodies. Am J Ophthal, 2005; 139:780-794

Wicklund MP, Kissel JT, et al(2001) Paraproteinemic neuropathy. Curr Treat Options Neurol, 2001; 3:147-156

Wierda W, O’Brien S, Wen S et al.(20005) Chemoimmunotherapy with fludarabine, cyclophosphamide, and rituximab for relapsed and refractory chronic lymphocytic leukemia. J Clin Oncol 2005; 23(18):4070-8.

Wongsaroj P, Kahwaji J, Vo A, Jordan SC. (2015) Modern approaches to incompatible kidney transplantation. World J Nephrol. 2015 Jul 6;4(3):354-62. doi: 10.5527/wjn.v4.i3.354. PMID: 26167458; PMCID: PMC4491925.

Zaja F(2002) Efficacy and safety of rituximab in type II mixed cryoglobulinemia. Blood, 2002; 101:3827-3834

Zaja F, Iacona I, Masolini P, et al.(2002) B-cell depletion with rituximab as treatment for immune hemolytic anemia and chronic thrombocytopenia. Haematologica, 2002; 87:189-195

Zamanian RT, Badesch D, Chung L, Domsic RT, Medsger T, Pinckney A, Keyes-Elstein L, D'Aveta C, Spychala M, White RJ, Hassoun PM, Torres F, Sweatt AJ, Molitor JA, Khanna D, Maecker H, Welch B, Goldmuntz E, Nicolls MR.(2021) Safety and Efficacy of B-Cell Depletion with Rituximab for the Treatment of Systemic Sclerosis-associated Pulmonary Arterial Hypertension: A Multicenter, Double-Blind, Randomized, Placebo-controlled Trial. Am J Respir Crit Care Med. 2021 Jul 15;204(2):209-221. doi: 10.1164/rccm.202009-3481OC. PMID: 33651671; PMCID: PMC8650794.

Zarkhin V, Li L, Kambham N, et al.(2008) A randomized, prospective trial of rituximab for acute rejection in pediatric renal transplantation. Am J Transplant, 2008; 8:2489-2490

Zecca M, Nobili B, Ramenghi U, et al.(2003) Rituximab for the treatment of refractory autoimmune hemolytic anemia in children Blood, 2003; 101:3857-3861

Zheng X, Pallera AM, Goodnough LT, et al(2003) Remission of chronic thrombotic thrombocytopenic purpura after treatment with cyclophosphamide and rituximab. Ann Int Med, 2003; 138:105-108

Zivkovic SA, Lacomis D, Lentzsch.(2009) Paraproteinemic neuropathy. Leuk Lymphoma, 2009; 50:1422-1433

Zojer N, Kirchbacher K, Vesely M, et al.(2006) Rituximab treatment provides no clinical benefit in patients with pretreated advanced multiple myeloma. Leuk Lymphoma, 2006; 47:1103-1109


Group specific policy will supersede this policy when applicable. This policy does not apply to the Wal-Mart Associates Group Health Plan participants or to the Tyson Group Health Plan participants.
CPT Codes Copyright © 2024 American Medical Association.