Sepantronium

Sepantronium Bromide (YM155) Enhances Response of Human B-Cell Non-Hodgkin Lymphoma to Rituximab

Aya Kita, Keisuke Mitsuoka, Naoki Kaneko, Mari Nakata, Kentaro Yamanaka,
Makoto Jitsuoka, Sosuke Miyoshi, Akihiro Noda, Masamichi Mori, Takahito Nakahara, and Masao Sasamata
Drug Discovery Research, Astellas Pharma Inc., Tsukuba, Japan
Received April 27, 2012; accepted July 10, 2012

ABSTRACT

In the treatment of B-cell non-Hodgkin lymphoma (B-NHL) rituximab improves long-term survival in combination with con- ventional chemotherapy. However, because the majority of pa- tients with B-NHL eventually relapse, the development of more effective therapies is needed. Here, we evaluated the antitu- mor effects of a combination treatment involving sepantro- nium bromide (YM155), a first-in-class survivin suppressant, and rituximab in B-NHL xenograft mice models. To deter- mine the efficacy of the combination treatment, YM155- and rituximab-treated B-NHL cell xenografted mice were moni- tored for tumor size and survival and subjected to 2′-deoxy- 2′-18F-fluoro-D-glucose (18F-FDG) and 3′-18F-fluoro-3′- deoxythymidine (18F-FLT) positron emission tomography (PET) imaging. In addition, the cell proliferation status of ex- cised tumors was examined by Ki-67 immunohistochemistry. In DB, WSU-DLCL-2, and Mino xenograft-bearing mice, the com- bination treatment of YM155 and rituximab induced significant large B-cell lymphoma (DLBCL), grade 3 follicular lym- phoma, and mantle cell lymphoma. However, because many patients are not candidates for autologous stem cell transplant because of advanced age or comorbidities, a need exists for the development of novel therapeutic strat- egies to supplement current B-NHL treatment regimens.

Introduction

B-cell non-Hodgkin lymphoma (B-NHL) is a heterogeneous group of B-cell malignancies that display considerable ge- netic diversity, which reflects the differentiation status of lymphocytes at the time of transformation (Ku¨ ppers, 2005). The anti-CD20 antibody rituximab (Rituxan; Chugai Phar- maceutical Co. Ltd., Tokyo, Japan) is standard therapy for many types of CD20-positive B-cell lymphomas and improves long-term survival in combination with conventional chemo- therapy (Cheson and Leonard, 2008). Because the majority of B-NHL patients eventually relapse (Coiffier, 2007), high- dose chemotherapy followed by autologous stem cell trans- plant is the accepted second-line treatment for diffuse tumor growth inhibition and tumor regression compared with either single agent. On day 3 after the initiation of treatment a significant decrease in both 18F-FDG and 18F-FLT tumor uptake from pretreatment levels was observed in combination treat- ment groups. The Ki-67 proliferation index was significantly decreased on day 3 in the xenograft models treated with com- bination treatment, suggesting that the combination of YM155 and rituximab reduced cell proliferation and glucose metabo- lism. Furthermore, compared with monotherapy, combination treatment prolonged survival times of severe combined immu- nodeficient mice with disseminated WSU-FSCCL and Jeko B-NHL tumors. Our findings demonstrate that YM155 and ritux- imab combination treatment enhances antitumor activity in B- NHL xenografts, and 18F-FLT and 18F-FDG PET imaging may allow the early functional evaluation of treatment responses in patients with B-NHL.

In patients with B-NHL, increased expression levels of the antiapoptotic protein survivin are typically observed in ma- lignant lymphocytes (Ambrosini et al., 1997). Survivin posi- tivity in patients with B-NHL is also associated with in- creased chemo-resistance and worse clinical outcomes (Adida et al., 2000; Markovic et al., 2011). In addition, functional knockdown of survivin by short hairpin RNA induces cell apoptosis and growth inhibition of B-NHL cells (Ansell et al., 2004). Taken together, these findings suggest that survivin-targeted therapy would be suitable for the treat- ment of B-NHL.

Sepantronium bromide (YM155) is a first-in-class com- pound that functions selectively to suppress survivin expres- sion (Nakahara et al., 2007). We demonstrated previously that YM155 displays potent anticancer activity against a broad spectrum of human cancer cell lines and various hu- man-derived tumor xenograft mouse models (Nakahara et al., 2011a; Yamanaka et al., 2011b). In aggressive B-NHL models, YM155 treatment resulted in potent and sustained antitumor activity (Kita et al., 2011). Immunohistochemical analyses of non-small cell lung cancer, melanoma, and B- NHL tumors have indicated that YM155-induced tumor re- gression is associated with the down-regulation of intratu- moral survivin expression, growth inhibition, and apoptotic induction of tumor cells (Kita et al., 2011; Nakahara et al., 2011b; Yamanaka et al., 2011a). Therefore, we hypothesized that YM155 treatment in combination with rituximab would be effective against B-NHL and evaluated the efficacy of combination treatment by using human B-NHL-xenografted mice models on tumor size and survival.

One powerful technique for evaluating the therapeutic ef- ficacy of drugs in the clinical setting is positron emission tomography (PET) imaging, which allows noninvasive as- sessment of functional and metabolic alterations in tumor cells. The glucose analog 2′-deoxy-2′-18F-fluoro-D-glucose (18F-FDG) is the most commonly used PET tracer for diag- nosis, staging, and response monitoring in patients with ma- lignant lymphoma including DLBCL (Klabbers et al., 2003; Shankar et al., 2006). A second attractive PET tracer is 3′-18F-fluoro-3′-deoxythymidine (18F-FLT), which is a sub- strate of the DNA synthetic pathway and considered to be highly specific for proliferating tumor cells (Shields et al., 1998; Vesselle et al., 2002). 18F-FLT uptake correlates with the rate of tumor proliferation, as measured by Ki-67 tumor staining, in patients with non-small cell lung cancer (Vesselle et al., 2002). However, the potential utility of 18F-FDG and
18F-FLT PET for the assessment of tumor response to YM155 treatment has not been demonstrated. Here, we evaluated the antitumor activity of YM155 combined with rituximab against B-NHL by interim 18F-FDG and 18F-FLT PET imag- ing using human B-NHL xenografted mice models.

Materials and Methods

YM155 and Rituximab. YM155 was synthesized in-house by Astellas Pharma, Inc. (Nakahara et al., 2007). For in vivo experi- ments, YM155 was dissolved and diluted in saline immediately be- fore administration. Dose levels are expressed in terms of YM155, the cationic moiety of the drug substance. Rituximab was obtained from Chugai Pharmaceutical Co., Ltd. (Tokyo, Japan) and diluted in saline immediately before administration.

Cell Lines. The human DLBCL cell line DB and the human mantle cell line Mino were purchased from the American Type Culture Collection (Manassas, VA). The human DLBCL cell line WSU-DLCL-2, human follicular lymphoma cell line WSU-FSCCL, and human mantle cell lymphoma cell line Jeko were purchased from the German Resource Center for Biological Material (Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, Braunschweig, Germany). The cells were maintained in RPMI 1640 medium (Invitrogen, Carlsbad, CA) supplemented with 10% heat-inactivated fetal bovine serum (Invitrogen) in a humidified incubator with an atmosphere containing 5% CO2 at 37°C. Animals for In Vivo Studies. All animal experimental proce- dures were approved by the institutional animal care and use com- mittee of Astellas Pharma, Inc., and Astellas Pharma, Inc., Tsukuba Research Center was awarded Accreditation Status by the Associa- tion for Assessment and Accreditation of Laboratory Animal Care International. Animals were euthanized if tumor burden exceeded 10% of the host animal’s body weight.

In Vivo Antitumor Activity in Subcutaneous Tumor Xeno- graft Models. Five-week-old male BALB/c nude mice (CAnN.Cg- Foxn1nu/CrlCrlj) were obtained from Charles River Japan (Kana- gawa, Japan). Cultured DB, WSU-DLCL-2, and Mino cells were harvested, suspended in a phosphate buffer solution, and then sub- cutaneously injected into the flanks of mice (3 × 106 cells/0.1 ml/ mouse). Body weight and tumor volume were assessed twice weekly. Tumor volume was estimated by calculating tumor dimensions using calipers (length × width2 × 0.5). When tumor volumes reached >300 mm3, animals were randomly assigned into groups based on tumor volume. In the combination treatment study, the first day of drug treatment was designated as day 0, and observations continued until day 21, representing one full treatment cycle in the clinical study of YM155. YM155 was administered as a 7-day continuous infusion by using a micro-osmotic pump (Alzet models 1003D and 1007D; Durect Corporation, Cupertino, CA). Rituximab was administered intrave- nously at 50 mg/kg on days 0 and 2. Percentage of tumor regression was calculated by using the following formula: 100 × [1 — (mean tumor volume of each group on day 21)/(mean tumor volume of each group on day 0)].

18F-FDG and 18F-FLT PET Imaging. 18F-FDG was purchased from Nihon Medi-Physics (Hakui, Japan). 18F-FLT was synthesized in-house from its precursors. In brief, 18F was produced via an 18O(p,n)18F nuclear reaction by proton bombardment (12 MeV; 40 µA) of an 18O-water target by using a cyclotron-target system (OSCAR-12; JFE Plant and Service Corp., Yokohama, Japan). 18F-FLT was then synthesized by nucleophilic fluorination of 3-N- boc-5′-O-dimethoxytrityl-3′-O-nosyl-thymidine. For PET imaging studies, 5 × 106 cells/0.1 ml/mouse were inoculated subcutaneously in the right hind leg. 18F-FDG and 18F-FLT uptake were evaluated by using a small-animal Inveon PET scanner (Siemens, Knoxville, TN) on days 0 and 3 of treatment. Selected mice for 18F-FDG PET scans were fasted for more than 12 h before imaging. For imaging, 6- to 12-MBq aliquots of 18F-FDG or 18F-FLT were administered by tail vein injection to conscious animals, which were then maintained in cages for 1 h to allow for 18F-FDG and 18F-FLT uptake into tumors. Under 2.0 to 2.5% isoflurane gas anesthesia, mice were placed on the scanning stage, and a 5-min emission scan was performed by using the PET scanner. Images were reconstructed by OSEM3D and ana- lyzed with ASIPro software (Seimens Medical Solutions, Malvern, PA). Regions of interest were drawn around the boundaries of tu- mors on axial slices, and the observed maximum pixel value was normalized to the injected dose and body weight to give the maxi- mum standardized uptake value (SUVmax).

Immunohistochemical Analysis of Lymphoma Xenografts. Tumors were excised from mice, weighed, fixed in 4% paraformalde- hyde, embedded in paraffin, and then sectioned at 5-µm thickness. The sections were dewaxed in xylene and then subjected to antigen retrieval by placing samples in 10 mM citrate buffer, followed by autoclaving for 15 min at 121°C. The sections were then incubated with mouse monoclonal anti-Ki-67 antibody (Dako North America, Inc., Carpinteria, CA) for 1 h. Antibody binding was visualized by using the Envision System (Madison, NJ). The sections were then incubated with diaminobenzidine and counterstained with hematox- ylin. The Ki-67 proliferation index of each group was calculated as follows: Ki-67-positive cells (per 1000 cells) × tumor weight (g).

Studies with Disseminated Xenografts. Four-week-old female SCID mice (CB17/Icr-Prkdcscid/CrlCrlj) were purchased from Charles River Japan. WSU-FSCCL and Jeko cells were injected intrave- nously (4 × 106 cells/0.2 ml/mouse), and dissemination and tumor growth were allowed to proceed for 7 days. The mice were then divided into four comparable groups of 10 animals each based on body weight by using SAS software (SAS Institute, Cary, NC), and a clinical dose of either YM155 (2 mg/kg 7-day continuous infusion), rituximab (50 mg/kg intravenously), or YM155 (2 mg/kg 7-day con- tinuous infusion) plus rituximab (50 mg/kg intravenously) was ad- ministered once every 3 weeks for three courses in WSU-FSCCL and one course in Jeko models. Animals were monitored daily for the occurrence of mortality and euthanized while under diethyl ether anesthesia. Mice were scored as dead if any of the following signs of suffering were observed: cachexia, weakening, and difficulty in mov- ing or eating.

Statistical Analysis. Differences in tumor volume on day 21 between the combination treatment group and each single-com- pound group were evaluated by using Student’s t test. Immunohis- tochemistry and PET imaging data were analyzed by using analysis of valiance followed by post hoc Tukey’s test. For survival analysis, Kaplan-Meier curves were generated, and the differences between the control and drug-treated groups were assessed by using the log-rank test. All statistical analyses were performed by using GraphPad Prism 5 (GraphPad Software, Inc., San Diego, CA), and P values less than 0.05 were considered to be significant. All values are expressed as means and S.E.M.

Results

YM155 Enhances Antitumor Activity of Rituximab against Subcutaneous Xenograft Tumors. We first eval- uated the antitumor effects of YM155 in combination with rituximab in B-NHL xenograft-bearing mice models (Fig. 1). Because our preliminary studies demonstrated that a 2 mg/kg dose of YM155 induced tumor regression, including a complete response, a dose lower than 2 mg/kg was used in the present study. In each xenograft model, rituximab mono- therapy showed slight antitumor activity. For YM155 mono- therapy, although a low dose of YM155 (0.5 or 1 mg/kg/day) caused a delay in tumor growth, tumors began to regrow during the cessation of treatment, and none of the tumors showed complete regression by day 21. In contrast, combina- tion treatment with YM155 and rituximab resulted in signif- icant tumor regression (>100% inhibition). The percentage of tumor regression in the combination treatment group on day 21 was 76, 29, and 9% for the DB-, WSU-DLCL-2-, and Mino-xenografted mice models, respectively (Fig. 1). A com- plete response was achieved in two of five DB-xenografted mice. In addition, no animals in any group exhibited signif- icant body weight loss at any time point during treatment (data not shown).

18F-FDG and 18F-FLT PET Imaging of WSU-DLCL-2

Xenograft-Bearing Mice. To assess the functional changes of tumor xenografts by the combination treatment, we per- formed 18F-FDG and 18F-FLT PET imaging as indicators of glucose metabolism and cell proliferation, respectively. Rep- resentative 18F-FDG (Fig. 2A) and 18F-FLT (Fig. 2B) images of WSU-DLCL-2 xenograft-bearing mice taken at baseline (before treatment, day 0) and 3 days post-treatment are shown. A marked decrease of tumoral FDG and FLT uptake 3 days after the initiation of combination treatment was observed (Fig. 2). In contrast, FDG and FLT uptake re- mained high in the control and rituximab monotherapy mice. Tumoral uptake of both FDG or FLT into the xenografts of all mice were analyzed, and the results are summarized in Table 1. Within 3 days of treatment, a decrease (—42%) in 18F-FDG uptake was achieved in the combination treatment group (P < 0.05 versus control). YM155 monotherapy and combi- nation treatment significantly reduced the tumor uptake of 18F-FLT by —32 and —39%, respectively (P < 0.05; P < 0.01 versus control). The combination treatment group also decreased 18F-FLT uptake compared with the rituximab mono- therapy group (—36%) (P < 0.05). Histology and Ki-67 Immunohistochemistry. To ex- amine the feasibility of PET analysis as a complementary technique to immunohistochemistry in routine clinical appli- cations, we also analyzed tissue samples by using a standard Ki-67 immunostaining assay as an indicator of cell prolifer- ation status. Representative sections from treated and un- treated tumors are shown in Fig. 3. We found that sections obtained from untreated WSU-DLCL-2 xenotransplants showed high proliferative activity. In YM155 monotherapy (—46%) and rituximab monotherapy (—23%), and combina- tion-treated animals (—56%), a significant decrease in the Ki-67 proliferation index was detected on day 3 (Table 2; P < 0.01, versus control). A significant decrease was observed in combination treatment tumors compared with rituximab monotherapy (P < 0.01). YM155 in Combination with Rituximab against Dis- seminated Aggressive B-NHL Xenografts. Advanced B- NHL commonly develops clinically as disseminated disease. To examine the survival benefit of YM155 and rituximab combination treatment, we evaluated the effects of the clin- ical dosage of YM155 (2 mg/kg/day) alone or in combination with rituximab in the disseminated SCID/WSU-FSCCL and SCID/Jeko lymphoma models (Fig. 4). The administration of either YM155 or rituximab alone extended the median sur- vival from 35 to 60 and 42 days, respectively, in WSU- FSCCL-xenografted mice, and from 54 to 67 and 90 days, respectively, in Jeko-xenografted mice. It is noteworthy that the addition of YM155 to rituximab treatment significantly prolonged survival compared with the respective monothera- pies in both examined models. Discussion The present study clearly shows that the survivin-suppres- sant YM155, in combination with rituximab, results in sig- nificant improvements in tumor regression and survival in B-NHL xenograft models. We also demonstrated that the antitumor effect of combined YM155 and rituximab treatment is accompanied by decreases in tumor glucose metabo- lism and cell proliferation, as assessed by small-animal 18F- FDG and 18F-FLT PET imaging. In the DB, Mino, and WSU-DLCL-2 xenograft models, although rituximab alone exhibited slight antitumor activity, the combination of low doses of YM155 (0.5 or 1 mg/kg/day) with rituximab significantly improved tumor response com- pared with the monotherapy groups (Fig. 1). Advanced B- NHL commonly develops clinically as disseminated disease. Here, the combination of YM155 and rituximab also signifi- cantly prolonged the survival of disseminated B-NHL models compared with either drug alone (Fig. 4). Because increased survivin dependence for tumor growth is reported to be as- sociated with poor clinical outcomes, the combination of a survivin suppressant, YM155, and an anti-CD20 antibody may represent a potent strategy to treat B-NHL (Adida et al., 2000; Markovic et al., 2011). A number of studies have suggested that the combination of rituximab with various agents potentially improves their therapeutic potential (Di Gaetano et al., 2001; Ghetie et al., 2001; Alas et al., 2002; Chow et al., 2002; Emmanouilides et al., 2002; Boye et al., 2003; Jazirehi et al., 2003; Ackler et al., 2010). The mechanisms of action of rituximab are not fully understood and remain a matter of debate. Two immune- mediated mechanisms, antibody-dependent cell-mediated cy- totoxicity and complement-dependent cytotoxicity, have at- tention as possible processes linked to rituximab action. To investigate whether natural killer-dependent antibody-de- pendent cell-mediated cytotoxicity activity is important for enhancing the effectiveness of YM155 treatment in vivo, we evaluated the combination regimen in lymphoma xenografts established in NOD/SCID mice, which have impaired T/B cell lymphocyte development and natural killer cell function (Shultz et al., 1995). Combined rituximab and YM155 treat- ment resulted in a significant enhancement of antitumor activity over rituximab alone, a finding that is consistent with the results in BALB/c nude mice (data not shown). This result suggests that the therapeutic interaction between these two agents does not seem to arise from the antibody- mediated effecter functions of rituximab, and the rituximab- mediated inhibition of intracellular survival pathways is im- portant for enhancing the effectiveness of YM155 treatment in vivo. Mechanistically, rituximab has been reported to ab- rogate the intracellular signal transduction of survival path- ways involving p38, mitogen-associated protein kinase, and nuclear factor-nB, resulting in decreased Bcl-2 expression and sensitization to drug-induced apoptosis. Rituximab also up-regulates Raf kinase inhibitor protein, thus decreasing the activity of the extracellular signal-regulated kinase 1/2 pathway, leading to reduced Bcl-xL expression (Bonavida, 2007; Vega et al., 2011). Previously, we have demonstrated that concomitant treatment with YM155 enhanced chemo-sensitivity to Bcl-2 inhibitor in DLBCL (data not shown). We postulate that the anticancer effects of YM155 and rituximab observed here result from the simultaneous inhibition of different cell survival pathways, the survivin pathway by YM155, and the Bcl-2/Bcl-xL pathway by rituximab. How- ever, Western blot analysis revealed that rituximab did not suppress the expression of Bcl-2/Bcl-xL in the DLBCL cell line (data not shown). Thus, further investigations are required to identify the signaling pathways that are af- fected by the combined exposure to YM155 and rituximab. We explored whether 18F-FDG and 18F-FLT accumulation are sufficiently large to be quantitatively imaged with PET early in the treatment. The uptake of 18F-FDG and 18F-FLT was significantly decreased in the combination-treated group by day 3; on the other hand, no apparent decrease was observed in the rituximab monotherapy group (Fig. 2). Both PET imaging techniques could detect early responses to treatment based on tumor activity, with results that were consistent with tumor regression determined by caliper mea- surements (Figs. 1 and 2). Recent animal and preliminary clinical studies have suggested that 18F-FLT may be more sensitive than 18F-FDG for assessing tumor response (Troost et al., 2010; Kahraman et al., 2011). Our present findings indicate that 18F-FDG and 18F-FLT have comparable sensi- tivities for detecting early changes in tumor cell activity after combined YM155 and rituximab treatment in a B-NHL xeno- graft model (Fig. 2), indicating that the combination treat- ment induces suppression of glucose metabolism and cell proliferation. It is noteworthy that the significant decrease in 18F-FLT uptake observed here in combination-treated mice was also consistent with a decrease in Ki-67 immunostaining (Fig. 3). Thus, the similar trends observed between PET output metrics and the standard histological marker Ki-67 suggest that PET imaging can be used to monitor cellular proliferation in tumors. These results suggest that PET im- aging with 18F-FDG or 18F-FLT could evaluate the response of tumors to YM155 and rituximab treatment within several days of initiating combined treatment, because this would allow nonresponding tumors to be identified. In conclusion, combined YM155 and rituximab treatment has shown promising antitumor effects in B-NHL xenograft mice models. In addition, PET imaging data suggest that tumor responses to combination treatment of YM155 and rituximab can be monitored by using the tracer 18F-FLT with a similar sensitivity to that of 18F-FDG-PET. Based on the positive preclinical results of YM155 in B-NHL, a phase II open-label study of YM155 in combination with rituximab in relapsed patients with CD20-positive B-NHL was initiated and is currently underway. Acknowledgments We thank M. Yamada (Astellas Research Technology, Inc., Tokyo, Japan) for help with the survival study; T. Kimura (Astellas Re- search Technology, Inc.) and J. Mizusawa (KAC Co., Ltd., Kyoto, Japan) for help with PET imaging studies; and members of our project team at Astellas Pharma Global Development, Inc. for con- structive comments on the manuscript. Authorship Contributions Participated in research design: Kita, Mitsuoka, Miyoshi, and Sasamata. Conducted experiments: Kita, Mitsuoka, Kaneko, Nakata, Jit- suoka, and Miyoshi. Contributed new reagents or analytic tools: Jitsuoka. Performed data analysis: Kita, Mitsuoka, Kaneko, Nakata, Jit- suoka, and Noda. Wrote or contributed to the writing of the manuscript: Kita, Mit- suoka, Kaneko, Yamanaka, Jitsuoka, Noda, Mori, Nakahara, and Sasamata. References Ackler S, Mitten MJ, Foster K, Oleksijew A, Refici M, Tahir SK, Xiao Y, Tse C, Frost DJ, Fesik SW, et al. (2010) The Bcl-2 inhibitor ABT-263 enhances the response of multiple chemotherapeutic regimens in hematologic tumors in vivo. Cancer Che- mother Pharmacol 66:869 – 880. Adida C, Haioun C, Gaulard P, Lepage E, Morel P, Briere J, Dombret H, Reyes F, Diebold J, Gisselbrecht C, et al. (2000) Prognostic significance of survivin expres- sion in diffuse large B-cell lymphomas. Blood 96:1921–1925. Alas S, Ng CP, and Bonavida B (2002) Rituximab modifies the cisplatin- mitochondrial signaling pathway, resulting in apoptosis in cisplatin-resistant non- Hodgkin’s lymphoma. Clin Cancer Res 8:836 – 845. Ambrosini G, Adida C, and Altieri DC (1997) A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma. Nat Med 3:917–921. Ansell SM, Arendt BK, Grote DM, Jelinek DF, Novak AJ, Wellik LE, Remstein ED, Bennett CF, and Fielding A (2004) Inhibition of survivin expression suppresses the growth of aggressive non-Hodgkin’s lymphoma. Leukemia 18:616 – 623. Bonavida B (2007) Rituximab-induced inhibition of antiapoptotic cell survival path- ways: implications in chemo/immunoresistance, rituximab unresponsiveness, prognostic and novel therapeutic interventions. Oncogene 26:3629 –3636. Boye J, Elter T, and Engert A (2003) An overview of the current clinical use of the anti-CD20 monoclonal antibody rituximab. Ann Oncol 14:520 –535. Cheson BD and Leonard JP (2008) Monoclonal antibody therapy for B-cell non- Hodgkin’s lymphoma. N Engl J Med 359:613– 626. Chow KU, Sommerlad WD, Boehrer S, Schneider B, Seipelt G, Rummel MJ, Hoelzer D, Mitrou PS, and Weidmann E (2002) Anti-CD20 antibody (IDEC-C2B8, ritux- imab) enhances efficacy of cytotoxic drugs on neoplastic lymphocytes in vitro: role of cytokines, complement, and caspases. Haematologica 87:33– 43. Coiffier B (2007) Rituximab therapy in malignant lymphoma. Oncogene 26:3603– 3613. Di Gaetano N, Xiao Y, Erba E, Bassan R, Rambaldi A, Golay J, and Introna M (2001) Synergism between fludarabine and rituximab revealed in a follicular lymphoma cell line resistant to the cytotoxic activity of either drug alone. Br J Haematol 114:800 – 809. Emmanouilides C, Jazirehi AR, and Bonavida B (2002) Rituximab-mediated sensi- tization of B-non-Hodgkin’s lymphoma (NHL) to cytotoxicity induced by paclitaxel, gemcitabine, and vinorelbine. Cancer Biother Radiopharm 17:621– 630. imab (anti-CD20) selectively modifies Bcl-xL and apoptosis protease activating factor-1 (Apaf-1) expression and sensitizes human non-Hodgkin’s lymphoma B cell lines to paclitaxel-induced apoptosis. Mol Cancer Ther 2:1183–1193. Kahraman D, Scheffler M, Zander T, Nogova L, Lammertsma AA, Boellaard R, Neumaier B, Ullrich RT, Holstein A, Dietlein M, et al. (2011) Quantitative analysis of response to treatment with erlotinib in advanced non-small cell lung cancer using 18F-FDG and 3'-deoxy-3'-18F-fluorothymidine PET. J Nucl Med 52:1871– 1877. Kita A, Nakahara T, Yamanaka K, Nakano K, Nakata M, Mori M, Kaneko N, Koutoku H, Izumisawa N, and Sasamata M (2011) Antitumor effects of YM155, a novel survivin suppressant, against human aggressive non-Hodgkin lymphoma. Leuk Res 35:787–792. Klabbers BM, Lammertsma AA, and Slotman BJ (2003) The value of positron emission tomography for monitoring response to radiotherapy in head and neck cancer. Mol Imaging Biol 5:257–270. Ku¨ ppers R (2005) Mechanisms of B-cell lymphoma pathogenesis. Nat Rev Cancer 5:251–262. Markovic O, Marisavljevic D, Cemerikic V, Perunicic M, Savic S, Filipovic B, and Mihaljevic B (2011) Clinical and prognostic significance of apoptotic profile in patients with newly diagnosed nodal diffuse large B-cell lymphoma (DLBCL). Eur J Haematol 86:246 –255. Nakahara T, Kita A, Yamanaka K, Mori M, Amino N, Takeuchi M, Tominaga F, Kinoyama I, Matsuhisa A, Kudou M, et al. (2011a) Broad spectrum and potent antitumor activities of YM155, a novel small-molecule survivin suppressant, in a wide variety of human cancer cell lines and xenograft models. Cancer Sci 102: 614 – 621. Nakahara T, Takeuchi M, Kinoyama I, Minematsu T, Shirasuna K, Matsuhisa A, Kita A, Tominaga F, Yamanaka K, Kudoh M, et al. (2007) YM155, a novel small-molecule survivin suppressant, induces regression of established human hormone-refractory prostate tumor xenografts. Cancer Res 67:8014 – 8021. Nakahara T, Yamanaka K, Hatakeyama S, Kita A, Takeuchi M, Kinoyama I, Mat- suhisa A, Nakano K, Shishido T, Koutoku H, et al. (2011b) YM155, a novel survivin suppressant, enhances taxane-induced apoptosis and tumor regression in a human Calu 6 lung cancer xenograft model. Anticancer Drugs 22:454 – 462. Shankar LK, Hoffman JM, Bacharach S, Graham MM, Karp J, Lammertsma AA, Larson S, Mankoff DA, Siegel BA, Van den Abbeele A, et al. (2006) Consensus recommendations for the use of 18F-FDG PET as an indicator of therapeutic response in patients in National Cancer Institute Trials. J Nucl Med 47:1059 – 1066. Shields AF, Grierson JR, Dohmen BM, Machulla HJ, Stayanoff JC, Lawhorn-Crews JM, Obradovich JE, Muzik O, and Mangner TJ (1998) Imaging proliferation in vivo with [F-18]FLT and positron emission tomography. Nat Med 4:1334 –1336. Shultz LD, Schweitzer PA, Christianson SW, Gott B, Schweitzer IB, Tennent B, McKenna S, Mobraaten L, Rajan TV, and Greiner DL (1995) Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol 154:180 –191. Troost EG, Bussink J, Hoffmann AL, Boerman OC, Oyen WJ, and Kaanders JH (2010) 18F-FLT PET/CT for early response monitoring and dose escalation in oropharyngeal tumors. J Nucl Med 51:866 – 874. Vega MI, Baritaki S, Huerta-Yepez S, Martinez-Paniagua MA, and Bonavida B (2011) A potential mechanism of rituximab-induced inhibition of tumor growth through its sensitization to tumor necrosis factor-related apoptosis-inducing li- gand-expressing host cytotoxic cells. Leuk Lymphoma 52:108 –121. Vesselle H, Grierson J, Muzi M, Pugsley JM, Schmidt RA, Rabinowitz P, Peterson LM, Vallie`res E, and Wood DE (2002) In vivo validation of 3'deoxy-3'- [18F]fluorothymidine ([18F]FLT) as a proliferation imaging tracer in humans: correlation of [18F]FLT uptake by positron emission tomography with Ki-67 im- munohistochemistry and flow cytometry in human lung tumors. Clin Cancer Res 8:3315–3323. Yamanaka K, Nakahara T, Yamauchi T, Kita A, Takeuchi M, Kiyonaga F, Kaneko N, and Sasamata M (2011a) Antitumor activity of YM155, a selective small-molecule survivin suppressant, alone and in combination with docetaxel in human malig- nant melanoma models. Clin Cancer Res 17:5423–5431. Yamanaka K, Nakata M, Kaneko N, Fushiki H, Kita A, Nakahara T, Koutoku H, and Sasamata M (2011b) YM155, a selective survivin suppressant, inhibits tumor spread and prolongs survival in a spontaneous metastatic model of human triple negative breast cancer. Int J Oncol 39:569 –575.