Progressive multifocal leukoencephalopathy after ...

6 downloads 0 Views 175KB Size Report
John F. Seymour,6 Jacob Laubach,7 Susie D. Bawn,8 Leo I. Gordon,2,3 Jane N. Winter,2,3 Richard R. Furman,9. Julie M. Vose,10 .... prine, and romiplostim.
From www.bloodjournal.org at AZIENDA OSPEDALIERA PISANA on May 18, 2009. For personal use only. CLINICAL TRIALS AND OBSERVATIONS

Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the Research on Adverse Drug Events and Reports project Kenneth R. Carson,1 Andrew M. Evens,2,3 Elizabeth A. Richey,2 Thomas M. Habermann,4 Daniele Focosi,5 John F. Seymour,6 Jacob Laubach,7 Susie D. Bawn,8 Leo I. Gordon,2,3 Jane N. Winter,2,3 Richard R. Furman,9 Julie M. Vose,10 Andrew D. Zelenetz,9,11 Ronac Mamtani,9 Dennis W. Raisch,12 Gary W. Dorshimer,13 Steven T. Rosen,2,3 Kenji Muro,14 Numa R. Gottardi-Littell,15 Robert L. Talley,16 Oliver Sartor,17 David Green,2,3 Eugene O. Major,18 and Charles L. Bennett2,3,19 1Siteman

Comprehensive Cancer Center, Washington University School of Medicine, St Louis, MO; 2Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL; 3Robert H. Lurie Comprehensive Cancer Center, Chicago, IL; 4Department of Hematology, Mayo Clinic College of Medicine, Rochester, MN; 5Division of Hematology, University of Pisa, Pisa, Italy; 6Division of Haematology and Medical Oncology, Peter MacCallum Cancer Centre and University of Melbourne, East Melbourne, Australia; 7Dana-Farber Cancer Institute, Boston, MA; 8Division of Hematology/Oncology, Stanford University School of Medicine, CA; 9Division of Hematology/Oncology, New York Presbyterian Hospital-Cornell Medical Center, NY; 10Section of Hematology/Oncology, University of Nebraska Medical Center, Omaha; 11Division of Hematology/Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY; 12Veterans Administration Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, University of New Mexico, Albuquerque; 13Pennsylvania Hospital, University of Pennsylvania Health System, Philadelphia; Departments of 14Neurological Surgery and 15Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL; 16Kansas City Cancer Center, MO; 17Tulane Medical School, New Orleans, LA; 18Laboratory of Molecular Medicine and Neuroscience, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD; and 19Veterans Administration Center for the Management of Complex Chronic Conditions, Jesse Brown VAMC, Chicago, IL

Rituximab improves outcomes for persons with lymphoproliferative disorders and is increasingly used to treat immune-mediated illnesses. Recent reports describe 2 patients with systemic lupus erythematosus and 1 with rheumatoid arthritis who developed progressive multifocal leukoencephalopathy (PML) after rituximab treatment. We reviewed PML case descriptions among patients treated with rituximab from the Food and Drug Administration, the manufacturer, physicians, and a literature review from 1997

to 2008. Overall, 52 patients with lymphoproliferative disorders, 2 patients with systemic lupus erythematosus, 1 patient with rheumatoid arthritis, 1 patient with an idiopathic autoimmune pancytopenia, and 1 patient with immune thrombocytopenia developed PML after treatment with rituximab and other agents. Other treatments included hematopoietic stem cell transplantation (7 patients), purine analogs (26 patients), or alkylating agents (39 patients). One patient with an autoimmune hemolytic anemia developed

PML after treatment with corticosteroids and rituximab, and 1 patient with an autoimmune pancytopenia developed PML after treatment with corticosteroids, azathioprine, and rituximab. Median time from last rituximab dose to PML diagnosis was 5.5 months. Median time to death after PML diagnosis was 2.0 months. The case-fatality rate was 90%. Awareness is needed of the potential for PML among rituximab-treated persons. (Blood. 2009;113:4834-4840)

Introduction Progressive multifocal leukoencephalopathy (PML) is a rare demyelinating disease of the central nervous system that results from reactivation of latent JC polyoma virus (JCV). The disease was first described 50 years ago in patients with chronic lymphocytic leukemia and Hodgkin lymphoma.1 Up to 92% of the adult population is JCV-seropositive.2 PML typically occurs in persons with suppressed cellular immunity, particularly those with HIV infection.2 In clinical studies conducted by Koralnik et al, 80% of reported PML patients have AIDS, 13% have hematologic malignancies, 5% are transplant recipients, and 2% have chronic inflammatory diseases.3 Before the HIV epidemic, more than 60% of cases were seen in patients with lymphoproliferative disorders. The risk of PML in persons with hematologic malignancies is

estimated to be 0.07%, with the highest incidence (0.5%) being reported in persons with chronic lymphocytic leukemia.4,5 JC viral reactivation with PML is a rare complication recently reported among 7 patients after treatment with natalizumab, a monoclonal antibody that interferes with T-lymphocyte trafficking and intercellular adhesion. In 2005, 3 cases of natalizumab-associated PML were described. All 3 patients had received more than 2 years of natalizumab before PML was diagnosed.6-8 Since then, 5 additional multiple sclerosis patients with natalizumab monotherapy have developed PML.9,10 Rituximab treatment has been associated with viral infectious complications. In February 2006, 9 years after the drug received its initial Food and Drug Administration (FDA) approval, the labeling

Submitted October 30, 2008; accepted February 22, 2009. Prepublished online as Blood First Edition paper, March 5, 2009; DOI 10.1182/blood2008-10-186999.

International Conference on Malignant Lymphoma, Lugano, Switzerland, June 7, 2008.

Presented in part at the 49th Annual Meeting of the American Society of Hematology, Atlanta, GA, December 10, 2007; the American Society of Clinical Oncology Annual Meeting, Chicago, IL, June 3, 2008; and the Tenth

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked ‘‘advertisement’’ in accordance with 18 USC section 1734.

4834

BLOOD, 14 MAY 2009 䡠 VOLUME 113, NUMBER 20

From www.bloodjournal.org at AZIENDA OSPEDALIERA PISANA on May 18, 2009. For personal use only. BLOOD, 14 MAY 2009 䡠 VOLUME 113, NUMBER 20

for rituximab was changed to include information about patients with non-Hodgkin lymphoma (NHL) who had developed serious viral infections after treatment with the drug. Infections included hepatitis B, cytomegalovirus, herpes simplex virus, varicella zoster virus, West Nile virus, and JC virus.11 In 2006 and 2007, the FDA, the European Medicines Agency, the World Health Organization, and the manufacturer disseminated safety alerts describing 2 patients with systemic lupus erythematosus (SLE) who developed PML after treatment with rituximab and other immunosuppressive medications.12-15 In September 2008, the FDA and rituximab manufacturers issued “Dear Health Care Professional” letters describing a third patient with rheumatoid arthritis who died of PML 18 months after receiving rituximab, corticosteroids, and methotrexate therapy.16,17 Herein, we describe 52 patients with lymphoid malignancies, 2 patients with SLE, 1 patient with rheumatoid arthritis, 1 patient with idiopathic autoimmune pancytopenia, and 1 patient with immune thrombocytopenia purpura who developed PML after rituximab treatment.

Methods Cases were identified among rituximab-treated patients by clinicians from 12 cancer centers or academic hospitals (22 cases) or by reviewing FDA reports (11 cases), the manufacturer’s database (30 cases), and publications (18 cases; MeSH search terms: leukoencephalopathy, rituximab, immunosuppressed, lymphoma, and leukemia).18-31 The search covered the period from 1997, the date of the first FDA approval granted for rituximab, to December 31, 2008. Duplicate reports were identified based on age, sex, and underlying illness. Inclusion criteria were receipt of rituximab therapy before PML diagnosis or symptoms; PML confirmation based on histologic examination of brain tissue (histology-confirmed) or magnetic resonance imaging showing lesions consistent with a demyelinating process and documentation of cerebrospinal fluid (CSF) JCV DNA by polymerase chain reaction (PCR; laboratory-confirmed); and no evidence of HIV infection.32

PML CASES AFTER RITUXIMAB THERAPY

4835

trexate, a platinum-containing chemotherapy regimen, and a tumor necrosis factor-␣ inhibitor. A median of 6 rituximab doses (range, 1-28 doses) preceded PML diagnosis. Median time from first rituximab dose to PML diagnosis was 16.0 months (range, 1.0-90.0 months) and from last rituximab dose to PML diagnosis was 5.5 months (range, 0.3-66.0 months; Table 1). Presenting findings included confusion/disorientation (54% of patients), motor weakness/hemiparesis (33%), poor motor coordination (25%), speech changes (21%), or vision changes (18%). Symptoms progressed over weeks to months. The diagnosis was primarily confirmed by magnetic resonance imaging and JCV detection in the CSF (54%) and by brain biopsy or autopsy in the remaining patients. Quantitative T-cell studies, available for 14 patients, identified CD4 lymphopenia (CD4⫹ lymphocyte counts ⬍ 500 cells/␮L; 9 patients) or decreased CD4/CD8 ratios (9 patients; Table 2). Median duration between last rituximab dose and PML diagnosis was shorter among patients who had CD4⫹ lymphocyte counts ⬍ 500 cells/␮L (3 months vs 17 months for patients with CD4⫹ lymphocyte counts ⬎ 500 cells/␮L). One rituximab-treated patient who had not previously received a hematopoietic stem cell transplantation, purine analog, or alkylating agent had a normal CD4⫹ cell count and normal CD4/CD8 ratio. Paraffin-fixed bone marrow core specimens from 3 patients obtained before rituximab treatment had JCV detectable by PCR. The case-fatality rate was 90%: 100% among PML cases diagnosed within 3 months of the last rituximab dose versus 84% among PML cases diagnosed more than 3 months after the last rituximab dose. PML treatments included cytarabine or antiviral or immunologic therapies. Of 5 patients who did not die of PML, 2 received no therapy, 1 received cytarabine, another mirtazapine, and the last received cidofovir, donor lymphocyte infusions, cytarabine, and risperidone. These patients had residual neurologic deficits including motor aphasia, hemiparesis, and visual defects.

Discussion Results The median patient age was 61 years (range, 30-89 years). Underlying diagnoses included B-cell lymphoproliferative disorders (52 patients), SLE (2 patients), rheumatoid arthritis (1 patient), autoimmune pancytopenia (1 patient), and immune thrombocytopenic purpura (1 patient). Autoimmune hemolytic anemia had developed in 2 patients with B-cell lymphoproliferative disorders. Seven patients with lymphoproliferative disorders had received prior hematopoietic stem cell transplantation (3 allogeneic and 4 autologous). One patient had developed a postrenal transplantation lymphoproliferative disorder. Among 7 patients who had undergone prior hematopoietic stem cell transplantation, 14% had received purine analog therapy and 100% had received both alkylating agents and corticosteroids. Of 49 PML patients who had not undergone transplantation procedures, prior medications included purine analogs (46%), alkylating agents (81%), and corticosteroids (75%). One NHL patient with an autoimmune hemolytic anemia had previously received only corticosteroids and rituximab; 1 patient with an idiopathic autoimmune pancytopenia had previously received corticosteroids, azathioprine, and rituximab; and 1 patient with immune thrombocytopenic purpura had received corticosteroids, danazol, intravenous immunoglobulin, azathioprine, and romiplostim. Prior therapies for the 2 patients with SLE included corticosteroids and alkylating agents and for the patient with rheumatoid arthritis therapy included corticosteroids, metho-

This is the first large case series of PML cases occurring among HIV-negative patients treated with rituximab. Fifty-two of the patients had a diagnosis of hematologic malignancies and 5 had autoimmune illnesses. Presenting findings were primarily neuropsychiatric. All patients had received prior therapies that affect immune function, including alkylating agents, corticosteroids, purine analogs, or drugs to prevent allogeneic stem cell or solid organ graft rejection. The case-fatality rate was 90%. In interpreting our findings, several factors should be considered. The pathophysiology of rituximab-associated PML is unclear, particularly with respect to the role of rituximab. The mechanism underlying viral reactivation after rituximab treatment is probably more complex than simple B-cell depletion.11,33 Pre-B cells that harbor JCV in a latent state may be released into circulation to repopulate B-cell functions after differentiation. B lymphocytes may stimulate cellular immune responses, to both autoantigens and foreign antigens.29,34 Stasi et al demonstrated changes in T-lymphocyte cytokine profiles among immune thrombocytopenic purpura patients who responded to rituximab, suggesting changes in T-lymphocyte activity after B-lymphocyte depletion.35 A role for B lymphocytes in JCV immune responses is supported by JCV reactivation identification and PML in patients with congenital disorders of humoral immunity.36,37 Our findings also suggest that hematopoietic progenitor cells may be a site of viral latency38; 3 patients in our cohort had JCV detected in paraffin-fixed bone

From www.bloodjournal.org at AZIENDA OSPEDALIERA PISANA on May 18, 2009. For personal use only. 4836

BLOOD, 14 MAY 2009 䡠 VOLUME 113, NUMBER 20

CARSON et al

Table 1. Characteristics of 57 cases of PML after rituximab treatment Characteristic

All patients (n ⴝ 57)

Median age, y (range)

61 (30-89)

Male, %

43.8

Transplanted patients* (n ⴝ 8) 47.5 (30-60)

Nontransplanted patients (n ⴝ 49) 63 (44-89)

62.5

41.7

Indication for rituximab treatment Chronic lymphocytic leukemia, %

24.6

0

24.5

Follicular lymphoma, %

19.3

12.5

20.4

Diffuse large B-cell lymphoma, %

12.3

25.0

10.4

Non-Hodgkin lymphoma, %

17.5

25.0

16.3

Waldenstrom macroglobulinemia, %

5.3

0

6.3

Mantle cell lymphoma, %

3.5

12.5

2.0

Marginal zone B-cell lymphoma, %

3.5

12.5

2.0

Small lymphocytic lymphoma, %

3.5

0

4.1

Burkitt lymphoma, %

1.8

0

2.0

Autoimmune hemolytic anemia, %

3.5

12.5

2.0

Systemic lupus erythematosus, %

3.5

0

4.1

Rheumatoid arthritis, %

1.8

0

2.0

Immune thrombocytopenic purpura, %

1.8

0

2.0

Autoimmune pancytopenia, %

1.8

0

2.0

Corticosteroid, %

78.9

87.5

75.5

Cyclophosphamide, %

73.7

87.5

69.4

Vinca alkaloid, %

57.9

75.0

53.1

Purine nucleoside analog, %

45.6

12.5

51.0

Anthracycline, %

49.1

62.5

44.9

Chlorambucil, %

21.1

75.0

24.5

Etoposide, %

19.3

62.5

12.2

Platinum compound, %

17.5

62.5

10.2

Cytarabine, %

21.0

25.0

12.2

1.8

0

2.0

Confusion, mental status changes, %

54.4

37.5

57.1

Focal motor weakness, hemiparesis, %

33.3

37.5

30.6

Loss of motor coordination, %

24.6

25.0

24.5

Speech changes, %

21.1

37.5

18.4

Vision changes, %

17.5

12.5

18.4

MRI findings and JCV DNA in CSF, %

54.4

50.0

55.1

Brain biopsy, %

35.1

50.0

32.7

8.8

0

10.2

Survival†

10.4

25.0

6.5

Median rituximab doses (before diagnosis), (range)

6 (1-28)

4 (2-16)

6 (1-28)

Median first dose to PML diagnosis, mo (range)

16.0 (1.0-90.0)

24.0 (1.0-90.0)

14.0 (1.8-66.9)

Median last dose to PML diagnosis, mo (range)

5.5 (0.3-66.0)

6.0 (0.3-22.9)

5.5 (0.4-66.0)

Median PML diagnosis to death, mo (range)

2.0 (0.4-12.2)

2.0 (1.0-7.1)

2.5 (0.4-12.2)

Previous chemotherapy exposure

Alemtuzumab, % Presenting symptoms

Method of PML diagnosis

Autopsy, %

Median previous and concomitant chemotherapy

4 (1-14)

8 (3-10)

4 (1-14)

treatments (range) *Includes 1 patient who developed a lymphoproliferative disorder after a renal transplantation. †Survival information was available for 8 transplantation patients and 36 nontransplantation patients.

marrow samples obtained years before rituximab administration. Hematopoietic progenitor cells mobilized into the peripheral blood during chemotherapy may have been infected with latent JCV and may have facilitated hematogenous spread of JCV into the central nervous system. However, JCV latency may be more relevant for natalizumab-treated patients because CD34⫹ cells have been shown to mobilize into the peripheral circulation upon initiation of treatment.39-41 A high degree of awareness for PML facilitated case identification. This could account for the observation that most of the reported PML cases were from referral centers. At these centers, clinicians probably include PML in the differential diagnosis of hematologic malignancy patients who experience neuropsychiatric disturbances. One case of a rituximab-treated patient with rheumatoid arthritis was diagnosed as part of a long-term postmarketing safety study conducted by the manufacturer. It should be noted that

recognition of PML in the general medical community probably has improved recently because of the widely publicized association of PML with natalizumab use. A definitive diagnosis of PML can be based on compatible clinical findings, neuroimaging results, and characteristic histopathologic features with JCV detection in the brain tissue.42 However, brain biopsy is an invasive method with considerable risks. Others have proposed less invasive diagnostic methods based on amplification of the JCV-target DNA from CSF by PCR. Fong et al showed that the sensitivity and specificity of JCV DNA by PCR were 74% and 96%, respectively, and positive and negative predictive values were 89.5% and 88.5%, respectively (before the highly active antiretroviral therapy era).43 Persistent JC viremia in serum occurs in approximately 18% of HIV-positive patients without PML and 48% of HIV-infected patients with PML.44 Given the rarity of PML and the high incidence of transient JC viremia,

From www.bloodjournal.org at AZIENDA OSPEDALIERA PISANA on May 18, 2009. For personal use only. BLOOD, 14 MAY 2009 䡠 VOLUME 113, NUMBER 20

PML CASES AFTER RITUXIMAB THERAPY

4837

Table 2. T-lymphocyte studies for a subset of 14 rituximab-related PML cases Case

Indication

CD4ⴙ cells/␮L

1

Diffuse large B-cell lymphoma

68

2

Systemic lupus erythematosus

71

3

Non-Hodgkin lymphoma

4

Diffuse large B-cell lymphoma

5

CD8ⴙ cells/␮L

CD4/CD8 ratio

Purine analog exposure

Survival

NA

None

No

No

57

1.25

None

No

No

89

41

2.17

Autologous

No

No

94

243

0.39

None

No

No

Mantle cell lymphoma

110

310

0.35

None

No

No

6

Follicular lymphoma

152

532

0.29

None

No

No

7

Follicular lymphoma

234

1015

0.23

Autologous

No

No

8

Chronic lymphocytic leukemia

287

311

0.92

None

No

Yes

9

Mantle cell lymphoma

403

1309

0.31

Allogeneic

No

No

10

Marginal zone B-cell lymphoma

551

2596

0.21

Autologous

No

Yes

11

Autoimmune hemolytic anemia

562

382

1.47

None

No

No

12

Follicular lymphoma

570

1871

0.30

None

No

No

13

Autoimmune hemolytic anemia

1059

3453

0.31

Allogeneic

No

Yes

14

Waldenstrom macroglobulinemia

2100

6200

0.34

None

Yes

No

CD4⫹

The normal range is 500 to 1600 cells/␮L; the normal NA indicates not applicable.

NA

Hematopoietic transplantation

CD8⫹

range is 400 to 1100 cells/␮L; the normal CD4/CD8 ratio is 0.8 to 2.7.

the positive predictive value of this test as a PML screening tool for rituximab-treated patients with NHL or autoimmune diseases is probably low.44,45 Risk factors for development of PML include having CD4⫹ lymphocyte counts less than 200 cells/␮L among HIV-infected persons, whereas risk factors among HIV-negative persons have not been reported.46 One case report describes an HIV-seronegative patient with diffuse large B-cell lymphoma and an absolute CD4⫹ T-lymphocyte count of 68 cells/␮L who developed PML and pneumocystis pneumonia 3 months after rituximab-containing chemotherapy treatment.30 Of note, PML has been reported in HIVnegative persons who have CD4⫹ lymphocytes counts in excess of 200 cells/␮L.47 In our cohort, 9 of 14 rituximab-treated patients for whom CD4 cells were reported had CD4 cell levels less than 500 cells/␮L. Two rituximab-associated PML patients with higher CD4⫹ lymphocyte counts had underlying B-cell disorders. Our data suggest 2 possible rituximab-associated PML syndromes. One syndrome is associated with short intervals between last rituximab dose and PML diagnosis among patients with low CD4⫹ lymphocyte counts and occasionally low IgG levels. The second syndrome is associated with longer intervals between last rituximab dose and PML diagnosis among patients with higher CD4⫹ lymphocyte counts. Laszlo et al reported that CD4⫹ lymphocyte cell counts decreased to a median of 216 CD4⫹ T cells/␮L among 25 low-grade lymphoma patients who received rituximab and chlorambucil; 2 of these patients developed cutaneous herpes zoster infections, 1 developed reactivation of hepatitis B-viral infection, and none developed PML or other opportunistic infections.48 More information on CD4⫹ lymphocyte counts and immunoglobulin levels among rituximab-treated persons is needed. Historically, treatment of PML rarely has been successful in the absence of immune system reconstitution, as was the case in this report as well.49 Cytarabine has been the most commonly administered PML therapy, although clinical benefit is infrequent. Among HIV-infected patients, mean survival after a diagnosis of PML was only 0.4 years before the introduction of highly active antiretroviral therapy versus 1.8 years more recently.46 Factors associated with increased rates of PML survival include having CD4⫹ cell counts more than 50 cells/␮L at diagnosis among HIV-infected patients46 and among non–HIV-infected patients, after hematopoietic stem cell transplantation procedures where immune reconstitution frequently occurs.50 In our study, survival rates were 29% among the NHL patients who developed PML after hematopoietic stem cell transplantation procedures versus 7% among the NHL patients who

did not undergo hematopoietic stem cell transplantation procedures. Similarly, Garcia-Suarez reported survival rates of 38% among NHL patients who developed PML after hematopoietic stem cell transplantation procedures versus 10% for NHL patients who did not undergo hematopoietic stem cell transplantation procedures.50 With close monitoring and early discontinuation of natalizumab, 2 of the 5 most recently diagnosed patients with natalizumab-associated PML have recovered.51 With increasing concern over PML occurring among persons who receive rituximab or natalizumab antibody therapies, focused risk management efforts have been developed. For natalizumab, the manufacturer implemented a Risk Minimization Action Plan (RiskMAP) in the United States called Tysabri Outreach: Unified Commitment to Health (TOUCH). This program is designed to prospectively assess the risk of PML associated with natalizumab, minimize PML risk, minimize death and disability resulting from PML, and promote informed risk-benefit decisions.52,53 Risks of natalizumab are addressed through educational efforts describing risks of PML associated with natalizumab, restricted distribution, and mandatory registration of prescribing clinicians, pharmacists, infusion center staff, and patients. The FDA has advised healthcare professionals that natalizumab monotherapy may confer a lower risk of PML than when natalizumab is administered with other immunomodulatory medications.54 Prospective follow-up of 5000 natalizumab-treated patients facilitates epidemiologic assessments. Risk management related to rituximab-associated PML differs. Primary efforts involve educating physicians who prescribe rituximab and patients who receive the drug about PML.12,16,55 In addition, as part of an enhanced pharmacovigilance plan, the manufacturer will report to the FDA information on all rituximabtreated patients with PML and provide annual estimates of PML incidence rates among populations where cases have been reported. The risk management efforts do not include restricting rituximab distribution to certain prescribers as this would adversely affect drug access for large numbers of persons. It should be noted that risk-benefit assessments take into consideration observations that rituximab added to chemotherapy prolongs progression-free survival for persons with follicular non-Hodgkin lymphoma and prolongs survival and can be curative for patients with diffuse large B-cell lymphoma.56-60 Rituximab is proving to have benefit for a wide range of immunologic syndromes, including autoimmune hemolytic anemia, immune thrombocytopenic purpura, thrombotic thrombocytopenic purpura, and rheumatoid arthritis.56,61-64

From www.bloodjournal.org at AZIENDA OSPEDALIERA PISANA on May 18, 2009. For personal use only. 4838

BLOOD, 14 MAY 2009 䡠 VOLUME 113, NUMBER 20

CARSON et al

Queries of medical directors of lymphoma referral centers about cases of rituximab-associated PML facilitated obtaining unique detailed case descriptions for 22 cases of PML. Very few of these patients had been spontaneously reported previously to the FDA’s MedWatch program or to the manufacturer, and the completeness of the case reports was markedly greater in reports provided by these persons in comparison to reports submitted directly to the FDA or the manufacturer. Follow-up information was frequently provided by these clinicians in response to queries from investigators affiliated with the Research on Adverse Drug Events and Reports (RADAR) project. The RADAR project is an National Institutes of Health–funded, multidisciplinary team, led by a hematologist/oncologist/health services researcher, and consists of 25 core investigators with training in clinical pharmacology, pharmacy, epidemiology, statistics, internal medicine, and various medical subspecialties, including hematology, oncology, cardiology, infectious disease, and neurology.65 RADAR focuses on identification, evaluation, and dissemination of information describing rare and potentially fatal adverse drug events. As reported with our prior studies, close communication between clinicians and our pharmacovigilance project facilitates compilation of reports that are more complete than those reported spontaneously to the manufacturer or to the FDA.66 An important benefit of the collaboration was that RADAR, rather than the clinicians, responded to all follow-up queries from the FDA and the manufacturer. Some study limitations should be noted. Epidemiologic estimates of PML incidence are difficult to derive. In the general population, PML is estimated to occur at 1 case per 200 000 persons.67 Among HIV-infected populations, incidence rates decreased from 3.3 cases per 1000 person-years at risk in 1995 to 1996 to 1.3 cases per 1000 person-years at risk in 2000 to 2006, after the introduction of highly active antiretroviral therapy.46 Among persons with multiple sclerosis or Crohn disease, the estimated incidence of natalizumab-associated PML is 1 PML case per 1000 natalizumab-treated patients.52 For PML-associated with rituximab, Kavanaugh and Matteson reported 2 PML cases per 8000 rituximab-treated SLE patients.68 It is not possible to accurately estimate the incidence of rituximab-associated PML among persons with hematologic malignancies because of incomplete reporting of PML cases among rituximab-treated patients and incomplete data on the number of unique patients with lymphoid malignancies who have received rituximab. It should also be noted that the epidemiology of PML in the setting of lymphoid malignancies has changed over time. Before 1990, most PML cases occurred among persons with Hodgkin disease, whereas in recent years, with the development of purine analogs, hematopoietic stem cell transplantation procedures, and rituximab, most PML cases occur among non–HIV-infected persons with NHL or chronic lymphocytic leukemia.50 Consideration should be given to conducting epidemiologic studies to prospectively evaluate incidence rates and risk factors for PML among cohorts of rituximab-treated patients with NHL, autoimmune diseases, SLE, rheumatoid arthritis, and multiple sclerosis. Finally, causation assessment is more difficult when PML occurs among rituximab- versus natalizumab-treated persons because PML occurs in the absence of rituximab therapy among persons with NHL or autoimmune diseases, whereas it has not been reported

in the absence of natalizumab therapy among persons with multiple sclerosis or Crohn disease. In conclusion, rituximab administration may increase risks of developing PML, although the absolute risk of developing PML is probably low. As use of rituximab expands to diverse clinical settings, clinicians and patients should be aware of the potential for PML after rituximab therapy. Awareness and reporting of rituximab-associated PML cases to the FDA are essential to improve our understanding of risk factors, natural course, and alternative therapeutic approaches. Despite widespread public health advisories describing 2 patients with SLE and 1 patient with rheumatoid arthritis who were diagnosed with rituximabassociated PML, we identified 22 previously unreported cases associated with lymphoid malignancies and immunologically mediated cytopenias. Early diagnosis of PML will prompt efforts at immune reconstitution, which may be beneficial in improving survival rates. Finally, early diagnosis before irreversible neurologic damage has occurred will be crucial for evaluation of the efficacy of new antiviral treatments.

Acknowledgments The authors acknowledge the receipt of data from Genentech (South San Francisco, CA). This study was supported in part by National Cancer Institute grant 1R01 CA125077-01A1 (C.L.B.).

Authorship Contribution: K.R.C. conceptualized and designed the research, analyzed data, and wrote and edited manuscript; A.M.E. helped conceptualize and design the research, and edited the manuscript; E.A.R. managed and analyzed the data and wrote and edited the manuscript; T.M.H., J.M.V., S.D.B., R.R.F., J.N.W., J.L., R.M., K.M., and R.L.T. provided vital data for this research; D.F. participated in the conception and interpretation of data and edited the manuscript; J.F.S. participated in the conception, design, and editing of the manuscript; L.I.G. participated in the design of the study and editing of the manuscript; J.N.W. participated in the conception and design of the study and editing of the manuscript; A.D.Z. participated in the design of the study and interpretation of the data; D.W.R. participated in the analysis and interpretation of the data; G.W.D. contributed data and participated in the interpretation and analysis of the data; S.T.R. participated in the conception and design of the study; N.R.G.-L. participated in the design of the study and edited the manuscript; O.S. interpreted the data and edited the manuscript; D.G. and E.O.M. conceptualized and designed the study and edited the manuscript; and C.L.B. conceptualized, designed, and analyzed the study, and wrote and edited the manuscript. Conflict-of-interest disclosure: A.M.E. and S.T.R. serve on the Genentech speakers’ bureau. All other authors declare no competing financial interests. Correspondence: Charles L. Bennett, Northwestern University, 710 North Fairbanks Ct, Olson Pavilion, Suite 8-250, Chicago, IL 60611; e-mail: [email protected].

From www.bloodjournal.org at AZIENDA OSPEDALIERA PISANA on May 18, 2009. For personal use only. BLOOD, 14 MAY 2009 䡠 VOLUME 113, NUMBER 20

PML CASES AFTER RITUXIMAB THERAPY

4839

References 1. Astrom KE, Mancall EL, Richardson EP Jr. Progressive multifocal leuko-encephalopathy: a hitherto unrecognized complication of chronic lymphatic leukemia and Hodgkin disease. Brain. 1958;81:93-111. 2. Major EO, Amemiya K, Tornatore CS, Houff SA, Berger JR. Pathogenesis and molecular biology of progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating disease of the human brain. Clin Microbiol Rev. 1992;5:4973. 3. Koralnik IJ, Schellingerhout D, Frosch MP. Case records of the Massachusetts General Hospital. Weekly clinicopathological exercises (Case 142004): a 66-year-old man with progressive neurologic deficits. N Engl J Med. 2004;350:18821893. 4. Gonzalez H, Bolgert F, Camporo P, Leblond V. Progressive multifocal leukoencephalitis (PML) in 3 patients treated with standard-dose fludarabine (FAMP). Hematol Cell Ther. 1999;41:183-186. 5. Power C, Gladden JG, Halliday W, et al. AIDSand non-AIDS-related PML association with distinct p53 polymorphism. Neurology. 2000;54:743746. 6. Kleinschmidt-DeMasters BK, Tyler KL. Progressive multifocal leukoencephalopathy complicating treatment with natalizumab and interferon beta-1a for multiple sclerosis. N Engl J Med. 2005;353:369-374. 7. Langer-Gould A, Atlas SW, Green AJ, Bollen AW, Pelletier D. Progressive multifocal leukoencephalopathy in a patient treated with natalizumab. N Engl J Med. 2005;353:375-381. 8. Van Assche G, Van Ranst M, Sciot R, et al. Progressive multifocal leukoencephalopathy after natalizumab therapy for Crohn disease. N Engl J Med. 2005;353:362-368. 9. Biogen. Tysabri update, February 2009. http:// library.corporate-ir.net/library/14/148/148682/ items/325387/50A3DDB4-521D–4A7A-AA575B35B92DD671_biibTysabri20Feb09.pdf. Accessed March 2, 2009. 10. Hartung HP. New cases of progressive multifocal leukoencephalopathy after treatment with natalizumab. Lancet Neurol. 2009;8:28-31. 11. Aksoy S, Harputluoglu H, Kilickap S, et al. Rituximab-related viral infections in lymphoma patients. Leuk Lymphoma. 2007;48:1307-1312. 12. FDA. Rituximab. FDA Alert: Information for Healthcare Professionals, Rockville, MD, December 2006. http://www.fda.gov/cder/drug/ InfoSheets/HCP/rituximab.pdf. Accessed March 2, 2009. 13. EMEA. European Public Assessment Report: Mabthera (revision), 2007. http://www.emea. europa.eu/humandocs/PDFs/EPAR/Mabthera/ H-165-PI-en.pdf. Accessed March 2, 2009. 14. WHO. WHO Pharmaceuticals Newsletter, 2007. http://www.who.int/medicines/publications/ newsletter/PN_3_2007.pdf. Accessed March 2, 2009. 15. Rituximab (Rituxan) [package insert]. South San Francisco, CA: Genentech; 2006. 16. Rituximab (Rituxan) [package insert]. South San Francisco, CA: Genentech; 2008. 17. Genentech and Biogen. Important Drug Warning New Safety Information. 2008. http://www.fda. gov/medwAtch/safety/2008/rituxan_DHCP_ Final%209411700.pdf. Accessed March 2, 2009. 18. Baehring JM, Vives K, Bannykh S. Progressive multifocal leukoencephalopathy in a patient with marginal zone B-cell lymphoma. J Neurooncol. 2007;85:289-290. 19. Brito-Babapulle F, Moule S, Stewart W, et al. Progressive multifocal leukoencephalopathy (PML) following rituximab-combination chemotherapy treatment: British Society for Haematology An-

nual Conference Abstract. Br J Haematol. 2006; 133:36. 20. Darbesio A, Bertoldo E, Guana R, Geda C. Progressive mutlifocal leukoencephalopathy complicating follicular lymphoma: a possible role for chemotherapy and anti-CD20 treatment. 4th National Conference of Medical Oncology Abstract. Ann Oncol. 2002;13:94. 21. Freim Wahl SG, Folvik MR, Torp SH. Progressive multifocal leukoencephalopathy in a lymphoma patient with complete remission after treatment with cytostatics and rituximab: case report and review of the literature. Clin Neuropathol. 2007; 26:68-73.

36. Narula S, LaRosa DF, Kamoun M, Dalmau J, Levinson AI. Progressive multifocal leukoencephalopathy in a patient with common variable immunodeficiency and abnormal CD8⫹ T-cell subset distribution. Ann Allergy Asthma Immunol. 2007;98:483-489. 37. Teramoto T, Kaneko H, Funato M, et al. Progressive multifocal leukoencephalopathy in a patient with X-linked agammaglobulinemia. Scand J Infect Dis. 2003;35:909-910. 38. Sabath BF, Major EO. Traffic of JC virus from sites of initial infection to the brain: the path to progressive multifocal leukoencephalopathy. J Infect Dis. 2002;186[suppl 2]:S180-S186.

22. Goldberg SL, Pecora AL, Alter RS, et al. Unusual viral infections (progressive multifocal leukoencephalopathy and cytomegalovirus disease) after high-dose chemotherapy with autologous blood stem cell rescue and peritransplantation rituximab. Blood. 2002;99:1486-1488.

39. Bonig H, Wundes A, Chang KH, Lucas S, Papayannopoulou T. Increased numbers of circulating hematopoietic stem/progenitor cells are chronically maintained in patients treated with the CD49d blocking antibody natalizumab. Blood. 2008;111:3439-3441.

23. Harris HE. Progressive multifocal leucoencephalopathy in a patient with systemic lupus erythematosus treated with rituximab. Rheumatology (Oxf). 2008;47:224-225.

40. Lindberg RL, Achtnichts L, Hoffmann F, Kuhle J, Kappos L. Natalizumab alters transcriptional expression profiles of blood cell subpopulations of multiple sclerosis patients. J Neuroimmunol. 2008;194:153-164.

24. Hasan MM, Taylor P. Progressive multifocal leucoencephalopathy in a case of chronic lymphocytic leukemia. Br J Haematol. 2005;130:808. 25. Kranick SM, Mowry EM, Rosenfeld MR. Progressive multifocal leukoencephalopathy after rituximab in a case of non-Hodgkin lymphoma. Neurology. 2007;69:704-706. 26. Ng C, Slavin MA, Seymour JF. Progressive multifocal leukoencephalopathy complicating Waldenstrom macroglobulinaemia. Leuk Lymphoma. 2003;44:1819-1821.

41. Zohren F, Toutzaris D, Klarner V, Hartung HP, Kieseier B, Haas R. The monoclonal anti-VLA-4 antibody natalizumab mobilizes CD34⫹ hematopoietic progenitor cells in humans. Blood. 2008; 111:3893-3895. 42. Gibson PE, Gardner SD, Field AM. Use of a molecular probe for detecting JCV DNA directly in human brain material. J Med Virol. 1986;18:8795.

27. Pelosini M, Focosi D, Rita F, et al. Progressive multifocal leukoencephalopathy: report of 3 cases in HIV-negative hematological patients and review of literature. Ann Hematol. 2007;87:405-412.

43. Fong IW, Britton CB, Luinstra KE, Toma E, Mahony JB. Diagnostic value of detecting JC virus DNA in cerebrospinal fluid of patients with progressive multifocal leukoencephalopathy. J Clin Microbiol. 1995;33:484-486.

28. Rey J, Belmecheri N, Bouayed N, et al. JC papovavirus leukoencephalopathy after first line treatment with CHOP and rituximab. Haematologica. 2007;92:e101.

44. Dubois V, Moret H, Lafon ME, et al. Prevalence of JC virus viraemia in HIV-infected patients with or without neurological disorders: a prospective study. J Neurovirol. 1998;4:539-544.

29. Steurer M, Clausen J, Gotwald T, et al. Progressive multifocal leukoencephalopathy after allogeneic stem cell transplantation and posttransplantation rituximab. Transplantation. 2003;76:435436.

45. Delbue S, Guerini FR, Mancuso R, et al. JC virus viremia in interferon-beta-treated and untreated Italian multiple sclerosis patients and healthy controls. J Neurovirol. 2007;13:73-77.

30. Yokoyama H, Watanabe T, Maruyama D, Kim SW, Kobayashi Y, Tobinai K. Progressive multifocal leukoencephalopathy in a patient with B-cell lymphoma during rituximab-containing chemotherapy: case report and review of the literature. Int J Hematol. 2008;88:443-447. 31. Hopfinger G, Plessl A, Grisold W, et al. Progressive multifocal leukoencephalopathy after rituximab in a patient with relapsed follicular lymphoma and low IgG levels and a low CD4⫹ lymphocyte count. Leuk Lymphoma. 2008;49: 2367-2369. 32. Cinque P, Koralnik IJ, Clifford DB. The evolving face of human immunodeficiency virus-related progressive multifocal leukoencephalopathy: defining a consensus terminology. J Neurovirol. 2003;9[suppl 1]:88-92. 33. Weber F, Goldmann C, Kramer M, et al. Cellular and humoral immune response in progressive multifocal leukoencephalopathy. Ann Neurol. 2001;49:636-642. 34. Houff SA, Major EO, Katz DA, et al. Involvement of JC virus-infected mononuclear cells from the bone marrow and spleen in the pathogenesis of progressive multifocal leukoencephalopathy. N Engl J Med. 1988;318:301-305. 35. Stasi R, Del Poeta G, Stipa E, et al. Response to B-cell depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura. Blood. 2007;110:2924-2930.

46. Engsig FN, Hansen AB, Omland LH, et al. Incidence, clinical presentation, and outcome of progressive multifocal leukoencephalopathy in HIVinfected patients during the highly active antiretroviral therapy era: a nationwide cohort study. J Infect Dis. 2009;199:77-83. 47. Berger JR, Major EO. Progressive multifocal leukoencephalopathy. Semin Neurol. 1999;19:193200. 48. Laszlo D, Bassi S, Andreola G, et al. Peripheral T-lymphocyte subsets in patients treated with rituximab-chlorambucil combination therapy for indolent NHL. Ann Hematol. 2006;85:813-814. 49. Brooks BR, Walker DL. Progressive multifocal leukoencephalopathy. Neurol Clin. 1984;2:299313. 50. Garcia-Suarez J, de Miguel D, Krsnik I, Banas H, Arribas I, Burgaleta C. Changes in the natural history of progressive multifocal leukoencephalopathy in HIV-negative lymphoproliferative disorders: impact of novel therapies. Am J Hematol. 2005;80:271-281. 51. Winstein K. Biogen Cites New Setback for MS Drug. Wall Street Journal. December 16, 2008: D2. 52. Kappos L, Bates D, Hartung HP, et al. Natalizumab treatment for multiple sclerosis: recommendations for patient selection and monitoring. Lancet Neurol. 2007;6:431-441. 53. Wysowski D, Bozic C. Natalizumab’s Risk Minimization Action Plan and Risk Management Plan:

From www.bloodjournal.org at AZIENDA OSPEDALIERA PISANA on May 18, 2009. For personal use only. 4840

BLOOD, 14 MAY 2009 䡠 VOLUME 113, NUMBER 20

CARSON et al

Food and Drug Administration’s Peripheral and Central Nervous System Drug Advisory Committee meeting. Gaithersburg, MD, 2006. 54. FDA. FDA Alert: Information for Healthcare Professionals: Natalizumab Injection for Intraveneous Use; August 2008. http://www.fda.gov/ cder/drug/InfoSheets/HCP/natalizumab 2008HCP.htm. Accessed March 2, 2009. 55. Genentech. Medication Guide Rituxan (rituximab); 2008. http://www.gene.com/gene/ products/information/pdf/rituxan_med_guide.pdf. Accessed March 2, 2009. 56. Coiffier B, Lepage E, Briere J, et al. CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-Bcell lymphoma. N Engl J Med. 2002;346:235242. 57. Feugier P, Van Hoof A, Sebban C, et al. Longterm results of the R-CHOP study in the treatment of elderly patients with diffuse large B-cell lymphoma: a study by the Groupe d’Etude des Lymphomes de l’Adulte. J Clin Oncol. 2005;23: 4117-4126. 58. Marcus R, Imrie K, Solal-Celigny P, et al. Phase III study of R-CVP compared with cyclophosphamide, vincristine, and prednisone alone in pa-

tients with previously untreated advanced follicular lymphoma. J Clin Oncol. 2008;26:4579-4586. 59. Pfreundschuh M, Trumper L, Osterborg A, et al. CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial (MInT) Group. Lancet Oncol. 2006;7:379-391. 60. Fu K, Weisenburger DD, Choi WW, et al. Addition of rituximab to standard chemotherapy improves the survival of both the germinal center B-cell-like and non-germinal center B-cell-like subtypes of diffuse large B-cell lymphoma. J Clin Oncol. 2008;26:4587-4594. 61. Arnold DM, Dentali F, Crowther MA, et al. Systematic review: efficacy and safety of rituximab for adults with idiopathic thrombocytopenic purpura. Ann Intern Med. 2007;146:25-33. 62. Cohen SB, Emery P, Greenwald MW, et al. Rituximab for rheumatoid arthritis refractory to antitumor necrosis factor therapy: results of a multicenter, randomized, double-blind, placebocontrolled, phase III trial evaluating primary efficacy and safety at 24 weeks. Arthritis Rheum. 2006;54:2793-2806. 63. Hauser SL, Waubant E, Arnold DL, et al. B-cell

depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358:676688. 64. Ng KP, Cambridge G, Leandro MJ, Edwards JC, Ehrenstein M, Isenberg DA. B cell depletion therapy in systemic lupus erythematosus: longterm follow-up and predictors of response. Ann Rheum Dis. 2007;66:1259-1262. 65. Bennett CL, Nebeker JR, Lyons EA, et al. The Research on Adverse Drug Events and Reports (RADAR) project. JAMA. 2005;293:2131-2140. 66. Bennett CL, Nebeker JR, Yarnold PR, et al. Evaluation of serious adverse drug reactions: a proactive pharmacovigilance program (RADAR) vs safety activities conducted by the Food and Drug Administration and pharmaceutical manufacturers. Arch Intern Med. 2007;167:1041-1049. 67. Holman RC, Torok TJ, Belay ED, Janssen RS, Schonberger LB. Progressive multifocal leukoencephalopathy in the United States, 1979-1994: increased mortality associated with HIV infection. Neuroepidemiology. 1998;17:303-309. 68. Kavanaugh A, Matteson E. Hotline: Rituximab and Progressive Multifocal Leukoencephalopathy. American College of Rheumatology; 2008. http://www.rheumatology.org/publications/hotline/ 0107leuko.asp. Accessed March 2, 2009.