Rituximab (chimeric anti-CD20 monoclonal antibody) is widely employed in the treatment of patients with B cell non-Hodgkin lymphoma (NHL). This agent has activity in both indolent and aggressive disease, alone and in combination with chemotherapy. Unfortunately, however, many patients develop resistant disease. Ongoing efforts to improve outcomes include changes in dose and schedule, as well as the use of other biologic agents or antibodies that may enhance activity when administered together with rituximab. A relatively new focus is the development of engineered anti-CD20 antibodies that are optimized for their capability to mediate antibody-mediated cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC). Human or humanized structures have also been employed to potentially improve these attributes, as well as to improve on pharmacokinetics and immunogenicity. Other studies in NHL have clearly demonstrated that radiolabeled anti-CD20 antibodies (such as I-131 tositumomab and Y-90 ibritumomab tiuxetan) may be useful in relapsed and refractory disease, and have potential utility as part of initial treatment as well. Further studies of these modified anti-CD20 antibodies are ongoing in order to optimize their use for maximal clinical benefit.

CD20-directed monoclonal antibodies clearly demonstrate clinical efficacy in patients with B cell non-Hodgkin’s lymphoma (NHL). The chimeric anti-CD20 antibody rituximab was first studied in recurrent indolent NHL, and the “standard” regimen of 4 doses administered weekly (375 mg/m2) resulted in an objective response rate of 48% with a median time to progression (TTP) of just over 1 year.1 In diffuse large B cell lymphoma and in mantle cell lymphoma, response rates are in the 30% range.2,3 This degree of activity can be quite meaningful, and is associated with manageable toxicity, but unfortunately substantial numbers of patients do not respond and many relapse. Strategies to potentially improve efficacy include increases in dose (particularly through “maintenance” or “extended induction” schedules), as well as the addition of chemotherapy or novel biologic agents. More recently, several groups have engineered anti-CD20 antibodies to improve their ability to mediate antibody-mediated cellular cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC).

Additionally, antibody structures with human characteristics may potentially augment immune-based anti-tumor activity, improve pharmacokinetics, and reduce immunogenicity (given their similarity to the host). Finally, radiolabeled antibodies not only provide antibody-mediated effects but also deliver targeted radiation, which can overcome resistance and enhance efficacy. Recent data suggest that this approach can be useful in refractory disease and is also promising when used as part of initial treatment.

Evidence has suggested that prolonged exposure (beyond 4 weekly infusions) may improve efficacy of single agent rituximab therapy (as discussed fully by Dr. Ghielmini in this volume). Pharmacokinetic studies of data from the pivotal trial indicated that 4 doses of rituximab at 375 mg/m2 do not generally result in steady state serum concentrations and that the mean serum half-life after the fourth infusion is approximately 200 hours.1 Antibody clearance can be variable among patients, and better responses have generally been associated with higher serum rituximab levels. While significant efforts have been made in exploration of extended dosing regimens, retreatment at progression or during remission,4,10 it is possible that antibody modifications toward a human or humanized structure would result in more favorable pharmacokinetics and enhanced efficacy.

In order to optimize a therapeutic anti-cancer agent, however, it is critical to understand its dominant mechanisms of action and reasons for resistance. (See “Unique toxicities and resistance mechanisms associated with monoclonal antibody therapy” by Dr. Friedberg in this volume for a full discussion.) Despite the utility of rituximab, the critical pathways in its activity remain under debate. Many investigators believe that ADCC is an dominant part of the process, and this opinion is consistent with findings that polymorphisms in Fc receptors correlate with response.11,12 Byrd and colleagues suggested that rituximab-induced apoptosis is important in its activity in chronic lymphocytic leukemia (CLL) and that response can be correlated with caspase activation.13 Finally, the role of complement-mediated cytotoxicity (and the complement regulatory proteins CD55 and CD59) has been explored, and some studies support a role for this pathway, though this remains controversial.14,15 Currently, it appears that rituximab may have multiple potential mechanisms of action and that the involved pathways may relate at least in part to the histologic subtype of the tumor, host characteristics, and concurrent use of other agents in combination such as chemotherapy. Despite the lack of clarity, however, engineering of novel anti-CD20 antibodies to enhance their anti-tumor immune effects has progressed through preclinical studies and is currently in phase I and II clinical trials in lymphoma patients.

Teeling and colleagues characterized two novel human anti-CD20 antibodies (2F2 and 7D8) developed from human immunoglobulin (Ig) transgenic mice.16 Preclinical studies in the presence of complement demonstrated greater CDC than rituximab, presumably relating to slower “off rates” resulting in greater interaction with C1q and enhanced downstream complement activation. Additionally the fully human structure offers potentially favorable attributes with respect to immunogenicity and pharmacokinetics. From this work, the HuMax-CD20 human IgG1 monoclonal antibody (2F2) is undergoing evaluation in three phase I/II clinical trials (in follicular lymphoma, in CLL, and in rheumatoid arthritis) by Genmab A/S. Hagenbeek and colleagues recently reported on 40 patients with follicular lymphoma who were treated with this agent.17 Subjects were treated with 4 weekly infusions of 300–1000 mg following acetaminophen and diphenhydramine premedication. Median age was 59 years and patients had a median of 2 prior treatment regimens (range 1–5). Circulating B cells were depleted following therapy, and no dose-limiting toxicity was observed. The median half-life of circulating antibody after the fourth infusion was 342 hours. Twelve of 24 subjects demonstrated objective response, including 4 CR and 1 CR unconfirmed. These data suggest that this agent is well tolerated and is associated with meaningful clinical activity. Further studies are ongoing.

PRO70769 is a humanized IgG1 anti-CD20 monoclonal antibody undergoing evaluation by Genentech, Inc.18 Preclinical studies of a day 1 and day 15 intravenous infusion schedule in cynomolgus monkeys demonstrated circulating, splenic and lymphatic B cell depletion. While human clinical studies are currently focused in autoimmune disorders, it is conceivable that this agent might also have a role in the treatment of B cell malignancies.

IMMU-106 (hA20), also a humanized anti-CD20 monoclonal antibody, has been developed by Immunomedics, Inc. This agent shares the same human IgG framework as epratuzumab, a humanized anti-CD22 monoclonal antibody with clinical activity in B cell malignancies.19 Preclinical studies have shown that this agent can directly induce apoptosis of B cells and that it mediates both ADCC and CDC.20 Additionally, IMMU-106 has clinical activity in Raji-bearing SCID mouse tumor models, which is enhanced by the addition of epratuzumab. Clinical trials are ongoing with this agent in B cell lymphomas, and objective responses (including complete responses) have been observed. Interestingly, a report noted that IMMU-106 could be administered safely to and provide a response in a patient with resistant systemic lupus erythematosus and human anti-chimeric antibodies due to prior rituximab therapy (which was no longer effective).21 Though further study is clearly needed, this provocative report suggests that human or humanized antibodies may have utility in situations where rituximab is contraindicated.

Given that most patients with B cell lymphoma receive rituximab as part of their initial therapy and/or routinely at relapse, establishing the potential benefits of a novel anti-CD20 monoclonal antibody in lymphoproliferative disorders is a challenge. While a new anti-CD20 antibody might have some activity in rituximab-refractory patients, response rates would be expected to relate to whether there are shared mechanisms of resistance between rituximab and other antibodies. The lower response rates to single-agent rituximab in CML (relative to follicular lymphoma, where the bar is high) suggest that this setting may be particularly appropriate for the testing of new molecules. In contrast, some agents have been evaluated in “rituximab sensitive” patients, or at least those who would not be expected to be refractory based on a prior response. Results observed in this population, with mixed histologies and varied previous treatment regimens, are difficult to interpret. Ultimately, most new anti-CD20 antibodies will likely require comparative studies to single agent rituximab in B cell malignancies. Ironically, the success of rituximab in lymphoma and its relative lack of immunogenicity in this patient population appears to drive some developers of second generation anti-CD20 molecules toward rheumatologic diseases as a more enticing target, as the routine use of rituximab for treatment of autoimmunity may be less straightforward.22 

The addition of a radioisotope to a therapeutic monoclonal antibody can improve its activity through targeted radiation.23 Two radiolabeled anti-CD20 monoclonal antibodies are approved by the US Food and Drug Administration, yttrium-90 ibritumomab tiuxetan and iodine-131 tositumomab, for use in patients with recurrent low-grade or transformed low-grade lymphoma. The 1-week duration of therapy is generally welcomed by patients and is clearly shorter than alternative treatment options. Reasonably long-term follow-up has been obtained over approximately 10 years in some studies, and the toxicity profile has been generally well characterized. Principal side effects are hematologic and occur from approximately 4 until 8 weeks after treatment, due to radiation effects on the bone marrow. Incidence of neutropenic fever and the need for transfusional or hematopoietic growth factor support is generally low. Additionally, I-131 tositumomab has been associated with low rates of human anti-mouse antibody formation (HAMA) and hypothyroidism in approximately 10% of subjects.

An attractive feature of radioimmunotherapy (RIT) is that in some cases it can provide durable multi-year responses, even in patients with disease that is resistant to chemotherapy.24,25 Recent data have also suggested that these drugs can be effective in rituximab-refractory patients, with some patients having remissions for several years despite prior short responsiveness or unresponsiveness to previous unlabeled anti-CD20 therapy.26,27 Evidence suggests that RIT can commonly provide remissions that are longer than those observed with previous treatments, a phenomenon that contrasts with usual expectations with chemotherapy.24,28 Efforts to predict which patients with lymphoma will respond to RIT and which subjects are most likely to have a durable response (and therefore perhaps should be considered for early use of RIT) have not clearly identified a clear set of characteristics associated with a favorable outcome.

As RIT clearly has utility in patients with resistant disease, a logical next step is to employ it early in the course of treatment to attain potentially even better efficacy. A trial which evaluated the use of I-131 tositumomab in indolent or transformed NHL at first or second relapse resulted in a response rate of 76%, with a complete response (CR) rate of 49%.29 Overall duration of remission was a median of 1.3 years, and approximately 25% of subjects were in a continuous remission from 2.6+ to 5.2+ years after RIT at the time of the report. One might hypothesize that initial treatment could provide even better outcomes, as RIT would be administered prior to the evolution of chemoresistance and at a time of even greater bone marrow reserve. Kaminski and colleagues evaluated RIT with single agent I-131 tositumomab as initial therapy for follicular NHL.30 Seventy-six patients with advanced stage follicular lymphoma received a 1-week course of RIT and were observed for safety and efficacy. The overall response rate was 95%, including 75% complete responses, and 80% of Bcl-2 polymerase chain reaction-positive patients (at baseline) converted to negative after therapy. At over 5 years’ median follow-up, estimated progression-free survival was 6.1 years. Hematologic toxicity was acceptable, while HAMA formation (which is uncommon in relapsed patients in relation to their immunosuppressed condition) occurred in most subjects. The implications of HAMA formation are unclear, but it could possibly interfere with subsequent treatment with other murine antibodies and rituximab.

These interesting findings have led to additional studies which will ultimately help to determine the role of RIT as a component of initial therapy for indolent lymphoma. The Southwest Oncology Group (SWOG) conducted a trial of CHOP followed by I-131 tositumomab as initial treatment for follicular NHL in 90 subjects. Treatment was well tolerated, with an overall response rate of 90% (67% CR) and a 2-year progression-free survival of 81%.31 Fludarabine and CVP (cyclophosphamide, vincristine and prednisolone) chemotherapy have also been employed in studies by our group in sequence with I-131 tositumomab in follicular lymphoma with similarly promising results.32 Initial results from these trials suggest that the upfront chemotherapy plus RIT approach in these regimens may be more active in low- and intermediate-risk follicular lymphoma international prognostic index (FLIPI) subjects. Other studies with Y-90 ibritumomab tiuxetan as initial therapy in combination with either rituximab or with chemotherapy are also ongoing. Currently SWOG and Cancer and Leukemia Group B (CALGB) are performing a randomized trial of CHOP plus concurrent rituximab versus CHOP followed by I-131 tositumomab as initial therapy for follicular NHL. Other studies are evaluating the use of RIT in other clinical situations, including as a component of an autologous stem cell transplant approach, where promising results with poor-prognosis patients have been observed.33,36 

Data clearly suggest that CD20-directed RIT can be a useful treatment option for many patients with B cell lymphoma, yet relatively few patients appear to be routinely receiving it in practice. Logistical considerations (regarding coordination at a treatment site between medical oncology and nuclear medicine staff) will become less of an issue as centers gain experience with administering RIT. Concerns remain regarding the risk of long-term toxicity (including secondary leukemias) that can be associated with the use of radiation. While these risks appear low (and in the range of those observed with conventional modalities),37,38 longer term data with larger numbers of patients will be of value. A major question remains whether RIT should be used mainly in refractory, late-stage patients, in first or second relapse, or as part of initial treatment. In particular, patients with responsive disease but with short remissions can benefit from the opportunity for a durable response that can realistically occur following RIT. Additional support for the early use of monoclonal antibody-based treatments is provided by evidence suggesting that monoclonal antibody-based strategies (including RIT) may be improving overall survival.39 Preliminary data also demonstrate that RIT may have limited activity in relapsed and refractory aggressive lymphomas, such as diffuse large B cell and mantle cell subtypes.40,41 Sequential chemotherapy-RIT regimens are currently under evaluation as part of initial treatment for these histologies. Difficulty remains in clearly establishing the best settings for the use of RIT and will require randomized trials with long-term follow-up for clarification. Until comparative trial results are available, the choice of RIT among the array of new treatment options will remain a subjective decision for lymphoma clinicians and their patients.

Table 1.

Selected novel anti-CD20 antibodies in clinical development.

AgentClinical trial settingRef.
HuMax-CD20 (Genmab S/A) Follicular NHL, CLL Rheumatoid arthritis 16,17  
PRO70769 (Genentech, Inc) Rheumatoid arthritis 18  
IMMU-106 (hA20) (Immunomedics, Inc) NHL 20  
AgentClinical trial settingRef.
HuMax-CD20 (Genmab S/A) Follicular NHL, CLL Rheumatoid arthritis 16,17  
PRO70769 (Genentech, Inc) Rheumatoid arthritis 18  
IMMU-106 (hA20) (Immunomedics, Inc) NHL 20  
Table 2.

Settings where the use of radioimmunotherapy (RIT) is under evaluation in non-Hodgkin lymphoma (NHL).

  • Chemotherapy-refractory indolent and transformed NHL

  • Rituximab-refractory indolent and transformed NHL

  • Single agent initial follicular NHL therapy

  • Combination chemotherapy + RIT in upfront indolent NHL

  • Combination rituximab + RIT in upfront indolent NHL

  • Combination chemotherapy + RIT in upfront diffuse large B cell lymphoma

  • Combination chemotherapy + RIT in upfront mantle cell lymphoma

  • RIT + chemotherapy in autologous stem cell transplantation

 
  • Chemotherapy-refractory indolent and transformed NHL

  • Rituximab-refractory indolent and transformed NHL

  • Single agent initial follicular NHL therapy

  • Combination chemotherapy + RIT in upfront indolent NHL

  • Combination rituximab + RIT in upfront indolent NHL

  • Combination chemotherapy + RIT in upfront diffuse large B cell lymphoma

  • Combination chemotherapy + RIT in upfront mantle cell lymphoma

  • RIT + chemotherapy in autologous stem cell transplantation

 

Center for Lymphoma and Myeloma, Weill Medical College of Cornell University and New York Presbyterian Hospital

Supported in part by a K23 award from the National Institutes of Health (RR16814), and grants from the Lymphoma Research Foundation, the Dorothy Rodbell Cohen Foundation, and the Brian Rooney Fund of the Lymphoma Foundation.

1
McLaughlin P, Grillo-Lopez AJ, Link BK, et al. Rituximab chimeric anti-CD20 monoclonal antibody therapy for relapsed indolent lymphoma: half of patients respond to a four-dose treatment program.
J Clin Oncol
.
1998
;
16
:
2825
–2833.
2
Coiffier B, Haioun C, Ketterer N, et al. Rituximab (anti-CD20 monoclonal antibody) for the treatment of patients with relapsing or refractory aggressive lymphoma: a multicenter phase II study.
Blood
.
1998
;
92
(6):
1927
–1932.
3
Foran JM, Rohatiner AZ, Cunningham D, et al. European phase II study of rituximab (chimeric anti-CD20 monoclonal antibody) for patients with newly diagnosed mantle-cell lymphoma and previously treated mantle-cell lymphoma, immunocytoma, and small B-cell lymphocytic lymphoma.
J Clin Oncol
.
2000
;
18
(2):
317
–324.
4
Piro LD, White CA, Grillo-Lopez AJ, et al. Extended Rituximab (anti-CD20 monoclonal antibody) therapy for relapsed or refractory low-grade or follicular non-Hodgkin’s lymphoma.
Ann Oncol
.
1999
;
10
:
655
–661.
5
Davis TA, Grillo-Lopez AJ, White CA, et al. Rituximab anti-CD20 monoclonal antibody therapy in non-Hodgkin’s lymphoma: safety and efficacy of re-treatment.
J Clin Oncol
.
2000
;
18
:
3135
–3143.
6
Gordan LN, Grow WB, Pusateri A, et al. Phase II trial of individualized rituximab dosing for patients with CD20-positive lymphoproliferative disorders.
J Clin Oncol
.
2005
;
23
:
1056
–1058.
7
Hainsworth JD, Litchy S, Burris HA, 3rd, et al. Rituximab as first-line and maintenance therapy for patients with indolent non-Hodgkin’s lymphoma.
J Clin Oncol
.
2002
;
20
:
4261
–4267.
8
Hainsworth JD, Burris HA, 3rd, Morrissey LH, et al. Rituximab monoclonal antibody as initial systemic therapy for patients with low-grade non-Hodgkin lymphoma.
Blood
.
2000
;
95
:
3052
–3056.
9
Ghielmini S, Schmitz H, Cogliatts S, et al. Prolonged treatment with rituximab significantly improves event-free survival and response duration compared with the standard weekly x 4 schedule.
Blood
.
2004
;
103
:
4416
–4423.
10
Hainsworth JD, Litchy S, Shaffer DW, et al. Maximizing therapeutic benefit of rituximab: maintenance therapy versus retreatment at progression in patients with indolent non-Hodgkin’s lymphoma—a randomized phase II trial of the Minnie Pearl Cancer Research Network.
J Clin Oncol
.
2005
;
23
:
1088
–1095.
11
Clynes RA, Towers TL, Presta LG, Ravetch JV. Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targest.
Nat Med
.
2000
;
6
(4):
443
–446.
12
Cartron G, Dacheux L, Salles G, et al. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene.
Blood
.
2002
;
99
(3):
754
–758.
13
Byrd JC, Kitada S, Flinn IW, et al. The mechanism of tumor cell clearance by rituximab in vivo in patients with B-cell chronic lymphocytic leukemia: evidence of caspase activation and apoptosis induction.
Blood
.
2002
;
99
(3):
1038
–1043.
14
Golay J, Zaffroni L, Vaccari T, et al. Biologic response of B lymphoma cells to anti-CD20 monoclonal antibody rituximab in vitro: CD55 and CD59 regulate complement-mediated cell lysis.
Blood
.
2000
;
95
(12):
3900
–3908.
15
Weng WK, Levy R. Expression of complement inhibitors CD45, CD55, and CD59 on tumor cells does not predict clinical outcome after rituximab treatment in follicular non-Hodgkin lymphoma.
Blood
.
2001
;
98
(5):
1352
–1357.
16
Teeling JL, French RR, Cragg MS, et al. Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin lymphomas.
Blood
.
2004
;
104
(6):
1793
–1800.
17
Hagenbeek A, Plesner T, Walewski, et al. A novel fully human anti-CD20 monoclonal antibody, first clinical results from an ongoing phase I/II trial in patients with follicular non-Hodgkin’s lymphoma [abstract].
Ann Oncol
.
2005
;
16
(S5): abstract
114
.
18
Vugmeyster Y, Beyer J, Howell K, et al. Depletion of B cells by a humanized anti-CD20 antibody PRO70769 in Macaca facicularis.
J Immunother
.
2005
;
28
(3):
212
–219.
19
Leonard JP, Coleman M, Ketas JC, et al. Phase I/II trial of epratuzumab (humanized anti-CD22 antibody) in indolent non-Hodgkin’s lymphoma.
J Clin Oncol
.
2003
;
21
:
3051
–3059.
20
Stein R, Qu Z, Chen S, et al. Characterization of a new humanized anti-CD20 monoclonal antibody, IMMU-106, and its use in combination with the humanized anti-CD22 antibody epratuzumab, for the therapy of non-Hodgkin’s lymphoma.
Clin Cancer Res
.
2004
;
10
:
2868
–2878.
21
Tahir H, Rohrer J, Bhatia A, et al. Humanized anti-CD20 monoclonal antibody in the treatment of severe resistant systemic lupus erythematosus in a patient with antibodies against rituximab.
Rheumatology
.
2005
;
44
(4):
561
–562.
22
Looney RJ, Anolik JH, Campbell D, et al. B cell depletion as a novel treatment for systemic lupus erythematosus: a phase I/II dose-escalation trial of rituximab.
Arthritis Rheum
.
2004
;
50
(8):
2580
–2589.
23
Davis TA, Kaminski MS, Leonard JP, et al. The radioisotope contributes significantly to the activity of radioimmuno-therapy.
Clin Cancer Res
.
2004
;
10
:
7792
–7798.
24
Kaminski MS, Zelenetz AD, Press OW, et al. Pivotal study of iodine I 131 tositumomab for chemotherapy-refractory low-grade or transformed low-grade B-cell non-Hodgkin’s lymphomas.
J Clin Oncol
.
2001
;
19
:
3918
–3928.
25
Gordon LI, Molina A, Witzig T, et al. Durable responses after ibritumomab tiuxetan radioimmunotherapy for CD20+ B-cell lymphoma: long-term follow-up of a phase 1/2 study.
Blood
.
2004
;
103
:
4429
–4431.
26
Witzig TE, Flinn IW, Gordon LI, et al. Treatment with ibritumomab tiuxetan radioimmunotherapy in patients with rituximab-refractory follicular non-Hodgkin’s lymphoma.
J Clin Oncol
.
2002
;
20
:
3262
–3269.
27
Horning SJ, Younes A, Jain V, et al. Efficacy and safety of tositumomab and iodine-131 tositumomab (Bexxar) in B-cell lymphoma, progressive after rituximab.
J Clin Oncol
.
2005
;
23
:
712
–719.
28
Leonard JP, Zelenetz AD, Vose JM, Kaminski MS. Impact of course of therapy on response rate and duration with the Bexxar therapeutic regimen (tositumomab and iodine I 131 tositumomab) and with chemotherapy [abstract].
Blood
.
2004
;
104
: abstract
132
.
29
Davies AJ, Rohatiner AZ, Howell S, et al. Tositumomab and Iodine I 131 tositumomab for recurrent indolent and transformed B-cell non-Hodgkin’s lymphoma.
J Clin Oncol
.
2004
;
22
:
1469
–1479.
30
Kaminski MS, Tuck M, Estes J, et al. I-131 tositumomab therapy as initial treatment for follicular lymphoma.
N Engl J Med
.
2005
;
352
:
496
–498.
31
Press OW, Unger JM, Braziel RM, et al. A phase 2 trial of CHOP chemotherapy followed by tositumomab/iodine I 131 tositumomab for previously untreated follicular non-Hodgkin lymphoma: Southwest Oncology Group Protocol S9911.
Blood
.
2003
;
102
(5):
1606
–1612.
32
Leonard JP, Coleman M, Kaminski MS, et al. FLIPI predicts outcome in 65 patients with previously untreated indolent NHL who received Bexxar in combination with chemotherapy [abstract].
Ann Oncol
.
2005
;
16
(S5): abstract
63
.
33
Vose JM, Bierman PJ, Enke C, et al. Phase I trial of iodine-131 tositumomab with high-dose chemotherapy and autologous stem-cell transplantation for relapsed non-Hodgkin’s lymphoma.
J Clin Oncol
.
2005
;
23
:
461
–467.
34
Gopal AK, Gooley TA, Maloney DG, et al. High-dose radioimmunotherapy versus conventional high-dose therapy and autologous hematopoietic stem cell transplantation for relapsed follicular non-Hodgkin lymphoma: a multivariable cohort analysis.
Blood
.
2003
;
102
(7):
2351
–2357.
35
Winter J, Inwards D, Spies S, et al. Zevalin (90YZ) doses > 0.5 mCi/Kg may be combined with high-dose BEAM and autotransplant (ASCT) [abstract].
Ann Oncol
.
2005
;
16
(S5): abstract
215
.
36
Nademanee A, Krishnan A, Fung H, et al. Yttrium 90 (90Y) ibritumomab tiuxetan (Zevalin) in the treatment of non-Hodgkin’s lymphoma (NHL): incorporation into autologous stem cell transplantation (ASCT) regimens [abstract].
Ann Oncol
.
2005
;
16
(S 5): abstract
216
.
37
Witzig TE, White CA, Gordon LI, et al. Safety of yttrium-90 ibritumomab tiuxetan radioimmunotherapy for relapsed low-grade, follicular, or transformed non-Hodgkin’s lymphoma.
J Clin Oncol
.
2003
;
21
(7):
1263
–1270.
38
Bennett JM, Kaminski MS, Leonard JP, et al. Assessment of treatment-related myelodysplastic syndromes and acute myeloid leukemia in patients with non-Hodgkin lymphoma treated with tositumomab and iodine I131 tositumomab.
Blood
.
2005
;
105
(12):
4576
–4582.
39
Fisher RI, LeBlanc M, Press OW, et al. New treatment options have changed the natural history of follicular lymphoma [abstract].
Blood
.
2004
;
104
:
168a
(abstract 583).
40
Morschhauser F, Huglo D, Martinelli G, et al. Yttrium-90 ibritumomab tiuxetan (Zevalin) for patients with relapsed/refractory diffuse large B-cell lymphoma not appropriate for autologous stem cell transplantation: results of an open-label phase II trial [abstract].
Blood
.
2004
:
104
:
41a
(abstract 130).
41
Oki Y, Pro B, Delpassand E, et al. A phase II study of yttrium-90 (90Y) ibritumomab tiuxetan (Zevalin) for treatment of patients with relapsed and refractory mantle cell lymphoma (MCL) [abstract].
Blood
.
2004
;
104
:
720a
(abstract 2632).