libo zhang †, % baruchel · 30/12/2015  · when exposed to hypoxia overnight, a two to 65-fold...

35
1 Combined antitumor therapy with metronomic topotecan and hypoxia-activated prodrug, evofosfamide, in neuroblastoma and rhabdomyosarcoma preclinical models Libo Zhang *,† , Paula Marrano , Bing Wu *,† , Sushil Kumar *, † , Paul Thorner †, % , Sylvain Baruchel *,‡,§ * New Agent and Innovative Therapy Program, Department of Paediatric Laboratory Medicine, Division of Hematology and Oncology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada; % Department of Laboratory Medicine and Pathobiology, University of Toronto; § Institute of Medical Sciences, University of Toronto, Toronto, Canada Running title: Preclinical study of evofosfamide and topotecan Key words: neuroblastoma, rhabdomyosarcoma, tumor models, preclinical study, hypoxia Address all correspondence to: Sylvain Baruchel, New Agent and Innovative Therapy Program, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada M5G 1X8. Phone: 416-813-5977; Fax: 416-813-5327; E-mail: [email protected] Disclosure of Potential Conflicts of Interest: No potential conflicts of interest were disclosed by the authors on April 3, 2021. © 2015 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

Upload: others

Post on 21-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Combined antitumor therapy with metronomic topotecan and hypoxia-activated prodrug, evofosfamide, in neuroblastoma and rhabdomyosarcoma preclinical models

    Libo Zhang *,†, Paula Marrano †, Bing Wu *,†, Sushil Kumar *, †, Paul Thorner †, % , Sylvain

    Baruchel *,‡,§

    *New Agent and Innovative Therapy Program, †Department of Paediatric Laboratory Medicine, ‡

    Division of Hematology and Oncology, Department of Paediatrics, The Hospital for Sick

    Children, Toronto, Canada; %Department of Laboratory Medicine and Pathobiology, University

    of Toronto; § Institute of Medical Sciences, University of Toronto, Toronto, Canada

    Running title: Preclinical study of evofosfamide and topotecan

    Key words: neuroblastoma, rhabdomyosarcoma, tumor models, preclinical study, hypoxia

    Address all correspondence to: Sylvain Baruchel, New Agent and Innovative Therapy Program, The Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, Canada

    M5G 1X8. Phone: 416-813-5977; Fax: 416-813-5327; E-mail: [email protected]

    Disclosure of Potential Conflicts of Interest: No potential conflicts of interest were disclosed by the authors

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 2

    Translational Relevance

    Tumor hypoxia is known to correlate with increased malignancy, metastatic potential, drug

    resistance and poor patient prognosis in a number of tumor types. Targeting tumor hypoxia with

    hypoxia-activated prodrugs (HAP), such as evofosfamide, may improve cancer therapy. So far,

    the effect of evofosfamide has been studied in combination with cytotoxic agents in adult

    cancers, whereas the impact of evofosfamide on pediatric cancers has not been investigated. This

    study focuses on the preclinical evaluation of efficacy of evofosfamide and its combination with

    topotecan, a topoisomerase I inhibitor widely used in recurrent neuroblastoma and

    rhabdomyosarcoma. We observed that targeting tumor hypoxia with HAPs improves antitumor

    effects of topotecan in our tumor models, which provides a new therapeutic approach for the

    treatment of pediatric solid tumors. All data gathered from this study will help us build the

    rationale for future phase 1 clinical trials in treating patients with relapsed/ refractory

    neuroblastoma and rhabdomyosarcoma.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 3

    Abstract:

    Purpose: Tumor cells residing in tumor hypoxic zones are a major cause of drug resistance and

    tumor relapse. In this study, we investigated the efficacy of evofosfamide, a hypoxia-activated

    prodrug, and its combination with topotecan in neuroblastoma (NBL) and rhabdomyosarcoma

    (RMS) preclinical models.

    Experimental Design: NBL and RMS cells were tested in vitro to assess the effect of

    evofosfamide on cell proliferation, both as a single agent and in combination with topotecan. In

    vivo antitumor activity was evaluated in different xenograft models. Animal survival was studied

    with the NBL metastatic tumor model.

    Results: All tested cell lines showed response to evofosfamide under normoxic condition, but

    when exposed to hypoxia overnight, a two to 65-fold decrease of IC50 was observed. Adding

    topotecan to the evofosfamide treatment significantly increased cytotoxicity in vitro. In NBL

    xenograft models, single-agent evofosfamide treatment delayed tumor growth. Complete tumor

    regression was observed in the combined topotecan / evofosfamide treatment group after two-

    week treatment. Combined treatment also improved survival in a NBL metastatic model,

    compared to single-agent treatments. In RMS xenograft models, combined treatment was more

    effective than single agents. We also showed that evofosfamide mostly targeted tumor cells

    within hypoxic regions while topotecan was more effective to tumor cells in normoxic regions.

    Combined treatment induced tumor cell apoptosis in both normoxic and hypoxic regions.

    Conclusions: Evofosfamide shows antitumor effects in NBL and RMS xenografts. Compared to

    single-agent, evofosfamide / topotecan, combined therapy improves tumor response, delays

    tumor relapse and enhances animal survival in preclinical tumor models.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 4

    Introduction:

    Neuroblastoma (NBL) is the most common extra-cranial childhood malignancy, responsible for

    15% of all childhood cancer deaths (1). Despite intensive treatment protocols including

    megatherapy with hematopoietic stem cell transplantation and immunotherapy, 3-year disease-

    free survival is only about 50% for metastatic disease compared to 95% for localized tumors (2).

    Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma and the third most

    common extracranial solid tumor in children, with an annual incidence of 4-7 cases per million

    children under the age of 16 years (3). Prognosis of metastatic RMS remains poor despite

    aggressive therapies.

    There is strong evidence that tumor cells residing in the tumor hypoxic regions cause drug

    resistance and tumor relapse (4). Tumor hypoxic zones, which occur in tumors due to non-

    uniform and inefficient vasculature, hinder the accessibility of chemotherapeutics to tumor cells.

    Hypoxia can be a direct cause of therapeutic resistance due to limited blood supply and drug

    distribution (5). Hypoxia also inhibits tumor cell proliferation and induces cell cycle arrest,

    further enhancing chemo-resistance through regulating drug transporters, anti-apoptotic proteins

    and pro-angiogenic factors (6). Our previous studies have shown that tumor hypoxic

    microenvironment may serve as a niche for the highly tumorigenic fraction of tumor cells which

    exhibit the cancer stem cell features in a diverse group of solid tumor cell lines, including

    neuroblastoma, rhabdomyosarcoma, and small-cell lung carcinoma (7). We also found that

    hypoxia induces drug resistance to etoposide, melphalan, doxorubicin, and cyclophosphamide in

    neuroblastoma cells, which is mediated through an expanded autocrine loop between VEGF/Flt1

    and HIF-1 alpha expression (8). There is evidence that some cells in hypoxic regions remain

    alive in a dormant state for prolonged duration and become drug resistant since anticancer drugs

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 5

    preferentially target rapidly proliferating cells (6). Hypoxia-inducible factors also induce stem

    cell phenotype and suppress differentiation of NBL cells (9). In RMS, HIF-1 alpha, induced by

    hypoxia, upregulates anti-apoptotic proteins and glycolytic enzymes (10). HIF-1 alpha

    inactivation by the inhibition of PI3K/Akt pathway is reported to sensitize tumor cells to

    apoptosis in RMS (11). Moreover, HIF-1 alpha stabilization as the result of hypoxia increases

    VEGF expression, increases glycolysis and resistance to chemotherapy (12).

    Evofosfamide (previously known as TH-302), a hypoxia activated prodrug (HAP) has been

    designed to penetrate to hypoxic regions of tumors. Evofosfamide is reduced at the

    nitroimadazole site of the prodrug by intracellular reductases and when exposed to hypoxic

    conditions, leads to the release of the alkylating agent bromo-isophosphoramide mustard (Br-

    IPM). Br-IPM can then act as a DNA crosslinking agent at the tumor hypoxic region and may

    diffuse to adjacent normoxic regions via a bystander effect (13). Consequently, evofosfamide can

    target the malignant cells residing in hypoxic zones, while having little or no cytotoxic effect on

    the normal cells (14, 15). The anti-tumor efficacy of evofosfamide correlated with hypoxic

    fractions in tumor xenografts of lung cancer, melanoma, pancreatic cancer, renal cell carcinoma

    and hepatocellular carcinoma. In adult soft tissue sarcoma Phase-II trial, evofosfamide in

    combination with doxorubicin demonstrated 2% complete response (CR) and 34% partial

    response (PR) (16). As a single maintenance therapy, it had limited hematologic toxicity and

    cardiotoxicity, whereas with doxorubicin, it caused manageable hematological toxicity without

    aggravating cardiotoxicity. A phase-III study in advanced and metastatic adult soft tissue

    sarcoma is ongoing (NCT01440088). To date, no studies investigating the efficacy of

    evofosfamide in pediatric cancers have been conducted.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 6

    Topotecan, a topoisomerase-I inhibitor has shown activity against both NBL and RMS as a

    single agent (17). The combination of cyclophosphamide plus topotecan has been active in

    rhabdomyosarcoma, neuroblastoma, and Ewing sarcoma patients with recurrent or refractory

    disease who have not received topotecan previously (18-20). The combination of topotecan,

    cyclophosphamide, and etoposide is tolerable and effective in relapsed and newly diagnosed

    neuroblastoma in phase II clinical trials and upfront phase III clinical trials (18, 21). Single agent

    topotecan has also been found to be antiangiogenic in preclinical models of pediatric cancers (22,

    23). Despite initial response, tumors eventually acquire resistance to topotecan. Mechanisms of

    resistance include mutations in topoisomerase-I and involvement of transporters like BCRP, P-

    glycoprotein and Multidrug Resistance Associated Protein-Type IV (24-26).

    In this study, we selected the metronomic dose of topotecan for in vivo studies. Low Metronomic

    Dose (LDM) chemotherapy usually refers to administration of low dose of cytototoxic agent at

    close intervals without drug free breaks. LDM chemotherapy has lower acute toxicity due to

    lower exposure of the cytotoxic agents. It has been shown active in diverse tumor types,

    including metastatic disease, especially when combined with antiangiogenic drugs (27-30) . The

    availability of the oral topotecan, along with our previous studies combining metronomic

    topotecan with pazopanib in NBL preclinical models (23), suggest that oral metronomic

    topotecan may be an ideal candidate for pediatric solid tumors. Considering the limited effects

    on hypoxic tumors with conventional chemotherapy, hypoxia-targeting cytotoxic agents along

    with topotecan therapy may achieve greater antitumor activities. Therefore, in this study, we

    investigated the effect of combined therapy with topotecan and evofosfamide in NBL and RMS

    preclinical tumor models.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 7

    Materials and Methods

    Drugs and reagents

    Evofosfamide was provided by Threshold Pharmaceuticals Inc. Topotecan HCl was obtained

    from Selleck (S1231). Stock solutions were prepared in DMSO and aliquots were stored at

    −80°C. They were further diluted with culture media or saline for in vitro and in vivo studies,

    respectively.

    Cells and Cell Culture

    RH4, RH30, and RD rhabdomyosarcoma cells (31) were gifts from Dr. David Malkin (The

    Hospital for Sick Children, Toronto). LAN-5 (32), SK-N-BE(2) (33), BE(2)C (34), and SH-

    SY5Y (34) neuroblastoma cells were kindly provided by Dr. Herman Yeger (The Hospital for

    Sick Children, Toronto). CHLA-15, CHLA-20 and CHLA-90 (35) were obtained from the

    Children's Oncology Group (COG) Cell Culture and Xenograft Repository under a signed and

    approved Material Transfer Agreement. Cell line authentication was performed using short

    tandem repeats (STR) DNA profiling (Promega’s GenePrint® 10 System)(36) conducted by the

    Genetic Analysis Facility at the Centre for Applied Genomics of The Hospital for Sick Children

    (Toronto, Canada). The DNA (STR) profile for all cell lines match to the profile listed in the

    Children's Oncology Group (COG) STR Database (http://strdb.cogcell.org). RH4, RH30 and RD

    rhabdomyosarcoma cells were cultured in DMEM supplemented with 10% FBS. CHLA-15,

    CHLA-20 and CHLA-90 neuroblastoma cells were cultured in Iscove’s modified Dulbecco’s

    medium supplemented with 3 mM l-glutamine, insulin, and transferin 5 μg/ml each and 5 ng/ml

    selenous acid (ITS Culture Supplement; Collaborative Biomedical Products, Bedford, MA) and

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 8

    20% fetal bovine serum (FBS, complete medium). SK-N-BE(2) and SH-SY5Y neuroblastoma

    cells were cultured in AMEM with 10% FBS.

    Test Animals

    Non-obese diabetic-severe combined immunodeficiency (NOD/SCID) mice (Jackson laboratory,

    Maine) were used for xenograft mouse models at 4 weeks of age. The mice were housed in an

    isolated sterile containment facility. All animal studies were approved by Animal Care Committee

    at the Hospital for Sick Children (Animal Use Protocol#1000019356). During the study the mice

    were observed daily for possible adverse effects due to treatments. Morbidity signs of ill health,

    such as ruffled/thinning fur, abnormal behaviors or local erosion from the tumor, were noted and

    experiments terminated as indicated.

    Cell Viability Assay

    Cell viability was assessed by Alamar Blue assay as previously described (37). Alamar Blue cell

    proliferation assay is based on a reducing environment that indicates metabolically active cells.

    Briefly, exponentially growing tumor cells was seeded into 48-well plates at 1 × 105 cells per

    well and grown overnight prior to initiating treatment. On the day of the test, cells were exposed

    to increasing concentrations of evofosfamide and topotecan either as single agents or combined

    treatment for 72 hours. Alamar Blue was added to a final concentration of 5%, incubated for 4

    hours at 37oC and quantitated on a plate reader at 530/590 nm. IC50 was determined by

    GraphPad Prism software.

    We also evaluated drug efficacy under the condition of hypoxia. To simulate tumor hypoxia in

    vitro, after drug addition, the plates were incubated for 2 hours at 37 °C in an anaerobic chamber.

    The anaerobic chamber was evacuated and gassed with the hypoxic gas mixture (94% N2/5%

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 9

    CO2/1% O2) to create a hypoxic environment. After 2 hours or overnight exposure to hypoxic

    condition, cells were removed from the hypoxia chamber and further incubated for 3 days in

    standard tissue-culture incubator. Cell viability was assessed by Alamar Blue assay as described

    above.

    Mouse Xenograft Models

    Two NBL cell lines (CHLA-20 and SK-N-BE(2)) and two RMS cell lines (RH4 and RD) were

    used to establish murine models (38). Briefly, tumor cells were washed three times with HBSS

    before injection. Cell suspensions were mixed 1:1 with Matrigel (BD Bioscience). Subcutaneous

    xenografts were developed by injecting 1 x 106 tumor cells into the subcutaneous fat pad of

    NOD/SCID mice. Treatments commenced when the tumor sizes reached about 0.25cm3.

    Animals were randomized into four groups with 10 animals in each group: control;

    evofosfamide, topotecan as a single agent, and the combination of evofosfamide and topotecan.

    The doses of evofosfamide and topotecan were 50 mg/kg (qd × 5/wk, IP) and 1 mg/kg (qd ×

    5/wk by oral gavage), respectively. The injection or gavage volume was 200 µL. Control mice

    received the same volume of the vehicle (saline). Tumor growth was measured three times a

    week in two dimensions, using a digital caliper. Tumor volume was calculated as width2 × length

    ×0.5. When tumor volume reached 1.5cm3, mice were sacrificed, and tumors were dissected and

    weighed. Tumor growth curves were plotted with the relative tumor volumes at different time

    points. The relative tumor volume of each tumor is defined as the tumor volume divided by its

    initial volume. Potential drug toxicity was assessed in tumor-bearing mice by monitoring animal

    body weight, appetite, diarrhea, signs of animal distress, or any marked neurologic symptoms.

    Animal Survival Study

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 10

    SK-N-BE(2) cells were prepared and injected i.v. into the lateral tail vein (26-gauge needle, 1 x

    106 cells in 100µl total volume). Mice were randomized into four groups: control, evofosfamide,

    topotecan and the combination of evofosfamide and topotecan, with 8 mice in each group. All

    the treatments were initiated 14 days after tumor cell inoculation at the the same schedule and

    dosage as above. Animals were monitored for body weight and any signs of stress. Tumor-

    bearing mice were euthanized at the endpoint when there were signs of distress, including 20%

    of body weight loss, fur ruffling, rapid respiratory rate, hunched posture, reduced activity, and

    progressive ascites formation.

    Immunohistochemistry

    Double immunofluorescence staining was performed on 5 μm thick frozen sections. Tissue

    sections were fixed for 10 minutes in acetone and allow to air dry for 5 minutes. Endogenous

    biotin, biotin receptors and avidin sites were block with the Avidin/Biotin Blocking Kit (SP-

    2001, Vector Laboratories, Burlingame, CA). The tissue sections were incubated with rabbit

    anti-cleaved caspase-3 antibody (1:50; #9661, Cell Signalling, Beverly, MA) or rabbit anti-Ki67

    antibody (1:200; ab16667, Abcam, Toronto, Canada) for 1 hour at room temperature. Detection

    of the rabbit antibodies was performed by incubation with Texas Red goat anti-rabbit IgG

    antibody (1:200; TI-1000, Vector Laboratories,) for 30 minutes. Following the washing of the

    tissue sections the Mouse IgG1 anti-pimonidazole monoclonal antibody clone 4.3.11.3 (1:50;

    Hypoxyprobe, Inc., Burlington, MA) was added overnight at 4°C. This antibody was detected

    with Fluorescein horse anti-mouse IgG Antibody (1:200; #FI-2000, Vector Laboratories) for 30

    minutes. The tissue sections were washed and then mounted with Vectashield mounting medium

    with DAPI (H-1200, Vector Laboratories).

    Statistical Analysis

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 11

    Data from different experiments were presented as mean ± SD. For statistical analysis, Student's

    t test for independent means was used. A P value of

  • 12

    All these cell lines were exposed to increased concentrations of evofosfamide in vitro for 72

    hours. Under normoxic condition, all the tested lines responded to evofosfamide treatment in a

    dose-dependent manner, with IC50 values ranging from 4.6 to 151µM. When exposing NBL and

    RMS cells under hypoxic conditions (1% O2) for 2 hours, no significant IC50 change was

    documented. However, when tumor cells were exposed to hypoxia (1% O2) overnight, there was

    a two to 65-fold decrease of evofosfamide IC50 (Fig. 1A, 1B, Tab 1) with the IC50 values

    ranging from 0.07 to 21.9µM.

    Improved anti-proliferation effects of evofosfamide when combined with topotecan in vitro

    In our previous studies, we showed that maximal plasma concentration (Cmax) of topotecan was

    19.8 ng/ml with the metronomic oral dose of 1 mg/kg drug administration in NOD/SCID mouse

    model (23). In patients, Cmax of topotecan is 10.6 ± 4.4 ng/ ml with the oral dose of 2.3 mg /m2

    /day. Since the terminal half-life of oral topotecan (3.9 ± 1.0 h) (40) is significantly longer than

    evofosfamide (0.6 ± 0.1 h) (41), a constant concentration of topotecan (20 nM= 8.4 ng/ml) was

    selected for in vitro combined treatment.

    All selected tumor cell lines were treated in vitro with increasing concentrations of evofosfamide

    with or without the presence of topotecan (20 nM). The dose response curves were generated by

    Graphpad software and IC50 values were compared between IC50 concentrations of

    evofosfamide when tested alone and in combination with 20nM topotecan in neuroblastoma and

    rhabdomyosarcoma cell lines.As shown in Tables 1 and 2, with the presence of topotecan,

    evofosfamide induced cytotoxicity was significantly enhanced under overnight hypoxia as

    indicated by decreased IC50s in most tested tumor cell lines.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 13

    To understand whether the combination of the drugs was additive or synergistic, a Bliss analysis

    was performed with MacSynergy II software with 95% confidence limits. The effect of the

    combination was additive in all cell lines.

    Topotecan and evofosfamide delay tumor growth and enhance animal survival in NBL model

    After verifying in vitro activity of evofosfamide and topotecan, the in vivo effectiveness of

    topotecan / evofosfamide as single-agents or combine therapy was evaluated in CHLA-20 and

    SK-N-BE(2) xenograft models. Since both CHLA-20 and SK-N-BE(2) cell lines are derived

    from samples of patients who relapsed after chemotherapy, we use them to develop aggressive

    tumor models for rigorous screening of anti-cancer agents. All treatments commenced when the

    tumor sizes reached 0.25cm3. In both NBL models, after 2 weeks of treatment, tumor growth

    delay was observed with evofosfamide or topotecan as monotherapies (Fig. 2A). In SK-N-BE(2)

    xenograft model, combine regimen induced complete tumor regression, while topotecan induced

    partial tumor regression. When we compared the tumor sizes between these two arms, the

    difference is statistically significant (p

  • 14

    Treatment was well tolerated, with weight loss >10% observed in single-agent and combined

    treatment groups within the first 8 days but returned to baseline thereafter. No animal death was

    observed in any of the groups during the experimental period (Fig. 2C).

    In order to determine the antitumor activities of evofosfamide and topotecan on late-stage

    metastatic disease, we further assess animal survival in the SK-N-BE(2) intravenous metastatic

    model. Fourteen days after tumor cell inoculation, we initiated drug treatment with evofosfamide

    and topotecan as single agents or combined treatment. As shown in Fig. 2D, evofosfamide or

    topotecan treatment showed a substantial increase in animal survival compared to the control

    mice with a median survival of 22.5 days for control group, 25 days for evofosfamide group and

    36 days for topotecan group. However, treatment with a combination of evofosfamide and

    topotecan had the greatest impact on animal survival (p < 0.01) with a median survival of 46

    days.

    Topotecan and evofosfamide delays tumor growth in RMS models

    To test the antitumor activity of evofosfamide and topotecan in rhabdomyosarcoma (RMS), we

    chose an alveolar RMS cell line (RH4) and an embryonal RMS cell line (RD) to generate

    xenograft models. Tumor bearing NOD/SCID mice were randomized to therapy with

    evofosfamide, topotecan, or combined therapy. In both RH4 and RD xenograft models, both

    evofosfamide and topotecan monotherapy significantly delayed tumor growth with 2 weeks of

    treatment (Fig. 3). Compared to NBL models, single-agent evofosfamide was more effective in

    RMS models with partial tumor regression in all treated xenografts. The combination of

    topotecan and evofosfamide were superior to the respective drugs given alone at different time

    points from day 26 to day 30 (P < 0.01; Fig. 3). There was no significant drug-related toxicity in

    any of the treatment arms.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 15

    Colocalization of cell apoptosis and tumor hypoxia with evofosfamide and topotecan treatment

    As combine treatment with evofosfamide and topotecan significantly improved anti-tumor

    activity both in vitro and in vivo, we have particular interest to examine its potential mechanism

    of action in vivo. To this end, tumor cell apoptosis was assessed by cleaved caspase-3

    immunoreactivity analysis in our xenograft models. On day 5 of drug treatment, RH4 xenografts

    from different regimens were harvested for immunohistochemical analysis (Fig. 4A). Hypoxic

    regions were identified as the area of pimonidazole positive staining. Apoptotic cells were

    detected by anti-cleaved caspase-3 antibody. Apoptosis was quantified as the percentage of

    positive cleaved caspase-3 staining area using ImageJ software (Fig. 4B). As shown in Fig. 4A

    and 4B, evofosfamide induced tumor cell apoptosis in hypoxic regions, with a remarkably higher

    percentage of cleaved caspase-3 positive cells in hypoxic regions compared to normoxic regions

    (Student's t test, p < 0.01). On the contrary, topotecan induced more extensive cell apoptosis in

    normoxic regions than hypoxic regions (p < 0.05). With the combination treatment, significant

    tumor cell apoptosis was observed throughout the tumor, in both normoxic and hypoxic areas.

    Decreased cell proliferation with evofosfamide and topotecan combined treatment in vivo

    We further evaluated tumor cell proliferation in different treatment groups by immunostaining of

    the proliferation marker, Ki67. As shown in Fig. 4C and 4D, the Ki67+ proliferating tumor cell

    population was mainly located in normoxic regions. After the 4-day in vivo treatment, single-

    agent topotecan showed a limited effect on cell proliferation as measured by Ki67 positivity.

    Surprisingly, decreased cell proliferation was observed in evofosfamide-treated tumors in

    normoxic regions with significantly lower percentage of Ki67 positive cells than control tumors

    (p < 0.05), suggesting a hypoxia-independent bystander effect. Combined evofosfamide and

    topotecan treatment led to a further decrease in the Ki67+proliferating tumor cell population

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 16

    compared to single-agent evofosfamide group (combined group vs. single-agent topotecan, p <

    0.01; combined group vs. single-agent evofosfamide, p < 0.05).

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 17

    Discussion

    Topotecan has demonstrated efficacy in preclinical and clinical studies in neuroblastoma and

    rhabdomyosarcoma. However, total eradication of highly tumorigenic populations of tumor cells

    is required to achieve relapse-free survival in cancer patients. Incomplete distribution of

    chemotherapeutics to poorly perfused areas of primary tumors and metastatic sites is a major

    hindrance in complete eradication of these tumor cells. These sparsely vascularized areas are

    characterized by hypoxia which confers drug and radiation resistance, and a pro-angiogenic and

    invasive phenotype to tumor cells. Despite the advent of novel chemotherapies, complete

    eradication of tumor cells cannot be accomplished until hypoxic tumor cells are targeted.

    Therefore, targeting hypoxic zone of solid tumor may reverse drug resistance and make cytotoxic

    agent more effective. In this study, we are proposing new strategies to improve the efficacy of

    chemotherapy by incorporating a novel agent targeting tumor hypoxic regions.

    Hypoxia activated prodrugs (HAP), which deliver the cytotoxic payload in hypoxic zones, have

    been developed to solve this issue. PR-104, an earlier HAP, was found to have a steep dose-

    response relationship in a panel of pediatric tumor xenograft models, which raises safety

    concerns in pediatric patients (42). Evofosfamide, however, has demonstrated survival benefit in

    various adult cancers without causing prohibitively additive toxicity with conventional

    chemotherapy in clinical trials (16). Evofosfamide shares the similar active metabolites as

    ifofosfamide, but it is preferentially activated in hypoxic tissues and does not have the same toxic

    metabolites as ifosfamide. It is anticipated that evofosfamide should have less hematologic, renal

    and CNS toxicity than ifosfamide. There is no direct clinical studies comparing evofosfamide

    with ifosfamide, but evofosfamide shows superior efficacy and less toxicity than ifosfamide in

    preclinical lung carcinoma models (43). In our xenograft models, we observed tumor growth

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 18

    inhibition with single agent evofosfamide, but no significant antitumor effect was achieved in the

    CHLA-20 model with ifosfamide (50 mg/kg; qd × 5/wk, IP) (Supplementary Figure 1). In

    clinical trials (NCT02047500), evofosfamide is administered at a dose ranging from 170-

    340 mg/m2. This could be converted to 32–64 mg/kg in the tumor bearing mice, calculated by the

    pharmacokinetic data from the clinical studies and preclinical mouse studies (44). In this study,

    we tested evofosfamide at the dose of 50mg/kg, as single agent and in combination with

    topotecan in two of the most common pediatric extracranial solid tumors, NBL and RMS. To our

    knowledge, this is the first study evaluating the effectiveness of evofosfamide in pediatric

    tumors.

    Our observation that single agent topotecan is more effective in delaying tumor growth in NBL

    than in RMS is in agreement with our previous finding (23). From our observations in vitro,

    evofosfamide showed more activity in neuroblastoma cell lines than in rhabdomyosarcoma cells.

    However, in vivo, rhabdomyosarcoma xenografts were more sensitive to evofosfamide than

    neuroblastoma xenografts. In this study, we can’t simply translate in vitro observation to in vivo

    activity. In vitro, we simulated tumor hypoxia by incubating tumor cells inside the hypoxia

    chamber. Therefore, 100% of cell population was under hypoxia and exposed to activated

    evofosfamide. In vivo, the percentage of hypoxic tumor cells varies among different tumor types

    (45, 46). In vivo tumor microenvironment is also more complex in which HAP activation would

    be affected by many other factors, including the duration and the intensity of oxidative stress,

    tumor angiogenesis, drug penetration in different solid tumor types, and acquired drug resistance

    under prolonged hypoxic conditions. Although it was beyond the scope of this study to examine

    the tumor microenvironment among different tumor types, it remains an important area for our

    future investigation.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 19

    From our study, evofosfamide showed enhanced cytotoxicity under overnight and 3-day

    (Supplementary Figure 2) prolonged hypoxia compared to short-term (2-hour) hypoxia

    exposure. When we test the effect of evofosfamide and its combination with topotecan on five

    neuroblastoma and three rhabdomyosarcoma cell lines under normoxia, short-term hypoxia and

    prolonged hypoxia, it is also under overnight hypoxia (1% O2) that adding topotecan

    significantly lowered IC50 of evofosfamide in most tumor cell line, except SK-N-BE(2).

    However, in the SK-N-BE(2) xenograft model, combined therapy achieved significant antitumor

    effects in vivo. Combined treatment induced complete tumor regression in all treated animals,

    which was superior to evofosfamide or topotecan single-agent treatment. This is likely because

    SK-N-BE(2) xenografts have more severe and more chronic hypoxic environment than in vitro

    cell culture (1% O2, overnight), which could more effectively activate evofosfamide to exert its

    cytotoxic activity in vivo.

    In all the NBL and RMS xenograft models tested, combined treatment showed superior

    antitumor effects compared to single-agent treatments, and induced tumor regression after a 2-

    week treatment period. The advantage of the combination regimen was also approved in the

    metastatic NBL model. Our experimental metastatic model has been developed to simulate

    residual disease in neuroblastoma. Residual Disease refers to disseminated tumor cells which

    survive after therapy (47). More than 50% of children with high-risk NBL develop recurrence

    due to the presence of minimal residual disease. In our metastatic model derived from a

    disseminated MYCN-amplified cell line, mice treated with combined topotecan and

    evofosfamide had extended survival compared to single-agent topotecan treatment. This result is

    significant in promoting new therapies for NBL minimal residual disease or refractory NBL by

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 20

    combining HAPs with existing chemotherapeutic drugs. We believe that incorporating HAPs,

    such as evofosfamide, could potentially eradicate the tumor cells hiding in hypoxic zones which

    are largely responsible for eventual relapse and metastasis. Eventually, this new regimen might

    prolong Progression Free Survival in high-risk NBL patients.

    In this study, we were able to assess the correlation between tumor hypoxia and tumor cell

    apoptosis with immunostaining of hypoxic marker pimonidazole and apoptoic marker cleaved

    caspase-3. As expected, in evofosfamide-treated tumors, apoptotic cells were mostly confined to

    hypoxic zones while those in topotecan-treated tumors were located in normoxic areas.

    Homogeneous population of tumor cells undergoing apoptosis was observed in both normoxic

    and hypoxic regions, which supported the mechanism of synergistic efficacy of the combination

    therapy in vivo. In a previous study, the accessibility of topotecan, doxorubicin and mitoxantrone

    was found to decrease with increasing distance from functional blood vessels in breast cancer

    xenografts (48). Clearly, poor drug penetration into solid tumors limits drug efficacy. In our

    study, we achieved tumor cytotoxicity in both normoxic and hypoxic regions by combining

    evofosfamide with topotecan, which explains the superior efficacy of our combination regimen

    in all tumor models.

    An ideal HAP requires selective activation in hypoxic regions as well as good penetration of

    multiple cell layers to exert a local cytotoxic bystander effect. Using a multicellular tumor

    spheroid and multicellular layer (MCL) co-culture model systems, Meng (49) et al. showed that

    evofosfamide has penetrability and bystander effect. In our study, with evofosfamide single-

    agent treatment, we observed decreased cell proliferation even at normoxic regions. This

    phenomenon confirms that once activated in hypoxic tissues, evofosfamide releases Br-IPM

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 21

    which can diffuse into surrounding oxygenated regions of the tumor and kill cells in those

    regions via a “bystander effect”.

    In summary, topotecan and evofosfamide demonstrated antitumor activities in our preclinical

    models of various sub-types of neuroblastoma and rhabdomyosarcoma. Compared to single

    agents, combination treatment induces more tumor regression, delays tumor relapse, and

    enhances animal survival in our preclinical tumor models. In this study, we explored the

    mechanism of action with combined evofosfamide/topotecan therapy. We observed increased

    reactive oxygen species (ROS) under hypoxic condition (1%O2, overnight) (Supplementary

    Figure 3). We also found that adding topotecan enhanced the oxidative stress in

    rhabdomyosarcoma cells under hypoxic condition (Supplementary Figure 4). In vivo,

    evofosfamide showed significant cytotoxicity against hypoxic tumor cells, while topotecan plays

    complementary roles by targeting tumor cells residing at normoxic regions. The ability of this

    combination to target cells in both normoxic and hypoxic tumor zones, as demonstrated by

    immunofluorescence, provides a proof of principle for its superior efficacy. These preclinical

    data support the development of a clinical trial in high-risk neuroblastoma and

    rhabdomyosarcoma.

    Acknowledgments This work was supported by Threshold Pharmaceuticals, Merck-Serono, James Birrell

    Neuroblastoma Research Fund and CJ Memorial Fund / SickKids Foundation.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 22

    References:

    1. Howman-Giles R, Shaw PJ, Uren RF, Chung DK. Neuroblastoma and other neuroendocrine tumors. Seminars in nuclear medicine. 2007;37:286-302. 2. Ora I, Eggert A. Progress in treatment and risk stratification of neuroblastoma: impact on future clinical and basic research. Semin Cancer Biol. 2011;21:217-28. 3. Huh WW, Skapek SX. Childhood rhabdomyosarcoma: new insight on biology and treatment. Curr Oncol Rep. 2010;12:402-10. 4. Vaupel P. The role of hypoxia-induced factors in tumor progression. The oncologist. 2004;9 Suppl 5:10-7. 5. Durand RE. The influence of microenvironmental factors during cancer therapy. In Vivo. 1994;8:691-702. 6. Brown JM. Tumor hypoxia, drug resistance, and metastases. J Natl Cancer Inst. 1990;82:338-9. 7. Das B, Tsuchida R, Malkin D, Koren G, Baruchel S, Yeger H. Hypoxia enhances tumor stemness by increasing the invasive and tumorigenic side population fraction. Stem Cells. 2008;26:1818-30. 8. Das B, Yeger H, Tsuchida R, Torkin R, Gee MF, Thorner PS, et al. A hypoxia-driven vascular endothelial growth factor/Flt1 autocrine loop interacts with hypoxia-inducible factor-1alpha through mitogen-activated protein kinase/extracellular signal-regulated kinase 1/2 pathway in neuroblastoma. Cancer Res. 2005;65:7267-75. 9. Jogi A, Ora I, Nilsson H, Lindeheim A, Makino Y, Poellinger L, et al. Hypoxia alters gene expression in human neuroblastoma cells toward an immature and neural crest-like phenotype. Proceedings of the National Academy of Sciences of the United States of America. 2002;99:7021-6. 10. Kilic M, Kasperczyk H, Fulda S, Debatin KM. Role of hypoxia inducible factor-1 alpha in modulation of apoptosis resistance. Oncogene. 2007;26:2027-38. 11. Kilic-Eren M, Boylu T, Tabor V. Targeting PI3K/Akt represses Hypoxia inducible factor-1alpha activation and sensitizes Rhabdomyosarcoma and Ewing's sarcoma cells for apoptosis. Cancer cell international. 2013;13:36. 12. Teicher BA. Hypoxia and drug resistance. Cancer metastasis reviews. 1994;13:139-68. 13. Duan JX, Jiao H, Kaizerman J, Stanton T, Evans JW, Lan L, et al. Potent and highly selective hypoxia-activated achiral phosphoramidate mustards as anticancer drugs. J Med Chem. 2008;51:2412-20. 14. Sun JD, Liu Q, Wang J, Ahluwalia D, Ferraro D, Wang Y, et al. Selective tumor hypoxia targeting by hypoxia-activated prodrug TH-302 inhibits tumor growth in preclinical models of cancer. Clin Cancer Res. 2011;18:758-70. 15. Liu Q, Sun JD, Wang J, Ahluwalia D, Baker AF, Cranmer LD, et al. TH-302, a hypoxia-activated prodrug with broad in vivo preclinical combination therapy efficacy: optimization of dosing regimens and schedules. Cancer Chemother Pharmacol. 2012;69:1487-98. 16. Chawla SP, Cranmer LD, Van Tine BA, Reed DR, Okuno SH, Butrynski JE, et al. Phase II study of the safety and antitumor activity of the hypoxia-activated prodrug TH-302 in combination with doxorubicin in patients with advanced soft tissue sarcoma. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2014;32:3299-306. 17. Nitschke R, Parkhurst J, Sullivan J, Harris MB, Bernstein M, Pratt C. Topotecan in pediatric patients with recurrent and progressive solid tumors: a Pediatric Oncology Group phase II study. Journal of pediatric hematology/oncology. 1998;20:315-8. 18. London WB, Frantz CN, Campbell LA, Seeger RC, Brumback BA, Cohn SL, et al. Phase II randomized comparison of topotecan plus cyclophosphamide versus topotecan alone in children with recurrent or refractory neuroblastoma: a Children's Oncology Group study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2010;28:3808-15.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 23

    19. Farhat R, Raad R, Khoury NJ, Feghaly J, Eid T, Muwakkit S, et al. Cyclophosphamide and Topotecan as First-line Salvage Therapy in Patients With Relapsed Ewing Sarcoma at a Single Institution. Journal of pediatric hematology/oncology. 2013. 20. Walterhouse DO, Lyden ER, Breitfeld PP, Qualman SJ, Wharam MD, Meyer WH. Efficacy of topotecan and cyclophosphamide given in a phase II window trial in children with newly diagnosed metastatic rhabdomyosarcoma: a Children's Oncology Group study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2004;22:1398-403. 21. Ashraf K, Shaikh F, Gibson P, Baruchel S, Irwin MS. Treatment with topotecan plus cyclophosphamide in children with first relapse of neuroblastoma. Pediatric blood & cancer. 2013;60:1636-41. 22. Soffer SZ, Kim E, Moore JT, Huang J, Yokoi A, Manley C, et al. Novel use of an established agent: Topotecan is anti-angiogenic in experimental Wilms tumor. Journal of pediatric surgery. 2001;36:1781-4. 23. Kumar S, Mokhtari RB, Sheikh R, Wu B, Zhang L, Xu P, et al. Metronomic oral topotecan with pazopanib is an active antiangiogenic regimen in mouse models of aggressive pediatric solid tumor. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17:5656-67. 24. Pan P, Li Y, Yu H, Sun H, Hou T. Molecular principle of topotecan resistance by topoisomerase I mutations through molecular modeling approaches. Journal of chemical information and modeling. 2013;53:997-1006. 25. Hendricks CB, Rowinsky EK, Grochow LB, Donehower RC, Kaufmann SH. Effect of P-glycoprotein expression on the accumulation and cytotoxicity of topotecan (SK&F 104864), a new camptothecin analogue. Cancer Res. 1992;52:2268-78. 26. Leggas M, Adachi M, Scheffer GL, Sun D, Wielinga P, Du G, et al. Mrp4 confers resistance to topotecan and protects the brain from chemotherapy. Molecular and cellular biology. 2004;24:7612-21. 27. Hashimoto K, Man S, Xu P, Cruz-Munoz W, Tang T, Kumar R, et al. Potent preclinical impact of metronomic low-dose oral topotecan combined with the antiangiogenic drug pazopanib for the treatment of ovarian cancer. Molecular cancer therapeutics. 2010;9:996-1006. 28. Merritt WM, Nick AM, Carroll AR, Lu C, Matsuo K, Dumble M, et al. Bridging the gap between cytotoxic and biologic therapy with metronomic topotecan and pazopanib in ovarian cancer. Molecular cancer therapeutics. 2010;9:985-95. 29. Minturn JE, Janss AJ, Fisher PG, Allen JC, Patti R, Phillips PC, et al. A phase II study of metronomic oral topotecan for recurrent childhood brain tumors. Pediatric blood & cancer. 2011;56:39-44. 30. Romiti A, Cox MC, Sarcina I, Di Rocco R, D'Antonio C, Barucca V, et al. Metronomic chemotherapy for cancer treatment: a decade of clinical studies. Cancer Chemother Pharmacol. 2013;72:13-33. 31. Rodriguez-Perales S, Martinez-Ramirez A, de Andres SA, Valle L, Urioste M, Benitez J, et al. Molecular cytogenetic characterization of rhabdomyosarcoma cell lines. Cancer genetics and cytogenetics. 2004;148:35-43. 32. Seeger RC, Siegel SE, Sidell N. Neuroblastoma: clinical perspectives, monoclonal antibodies, and retinoic acid. Annals of internal medicine. 1982;97:873-84. 33. Barnes EN, Biedler JL, Spengler BA, Lyser KM. The fine structure of continuous human neuroblastoma lines SK-N-SH, SK-N-BE(2), and SK-N-MC. In vitro. 1981;17:619-31. 34. Ciccarone V, Spengler BA, Meyers MB, Biedler JL, Ross RA. Phenotypic diversification in human neuroblastoma cells: expression of distinct neural crest lineages. Cancer Res. 1989;49:219-25. 35. Keshelava N, Seeger RC, Groshen S, Reynolds CP. Drug resistance patterns of human neuroblastoma cell lines derived from patients at different phases of therapy. Cancer Res. 1998;58:5396-405.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 24

    36. Masters JR, Thomson JA, Daly-Burns B, Reid YA, Dirks WG, Packer P, et al. Short tandem repeat profiling provides an international reference standard for human cell lines. Proceedings of the National Academy of Sciences of the United States of America. 2001;98:8012-7. 37. Zhang L, Smith KM, Chong AL, Stempak D, Yeger H, Marrano P, et al. In vivo antitumor and antimetastatic activity of sunitinib in preclinical neuroblastoma mouse model. Neoplasia. 2009;11:426-35. 38. Zhang L, Marrano P, Kumar S, Leadley M, Elias E, Thorner P, et al. Nab-paclitaxel is an active drug in preclinical model of pediatric solid tumors. Clin Cancer Res. 2013;19:5972-83. 39. Prichard MN, Shipman C, Jr. A three-dimensional model to analyze drug-drug interactions. Antiviral research. 1990;14:181-205. 40. Herben VM, Rosing H, ten Bokkel Huinink WW, van Zomeren DM, Batchelor D, Doyle E, et al. Oral topotecan: bioavailablity and effect of food co-administration. British journal of cancer. 1999;80:1380-6. 41. Ghobrial IM, Laubach JP, Raje N, Armand P, Schlossman RL, Boswell EN, et al. Phase 1 Study Of TH-302, An Investigational Hypoxia-Targeted Drug, and Dexamethasone In Patients With Relapsed/Refractory Multiple Myeloma. ASH 2013 Annual Meeting; 2013. 42. Houghton PJ, Lock R, Carol H, Morton CL, Phelps D, Gorlick R, et al. Initial testing of the hypoxia-activated prodrug PR-104 by the pediatric preclinical testing program. Pediatric blood & cancer. 2011;57:443-53. 43. Ahluwalia D SJ, Liu Q, Ferraro DJ, Wang Y, Lewis JG, et al. TH-302, a novel hypoxia-activated prodrug, shows superior efficacy and less toxicity than ifosfamide (IFOS) in metastatic and ectopic human lung carcinoma models. Proceedings of the AACR Annual Meeting. 2009;Apr 18–22; Denver, CO. Abstract 4517. 44. Jung D, Lin L, Jiao H, Cai X, Duan JX, Matteucci M. Pharmacokinetics of TH-302: a hypoxically activated prodrug of bromo-isophosphoramide mustard in mice, rats, dogs and monkeys. Cancer Chemother Pharmacol. 2012;69:643-54. 45. Reinhold HS, De Bree C. Tumour cure rate and cell survival of a transplantable rat rhabdomyosarcoma following x-irradiation. European journal of cancer. 1968;4:367-74. 46. Kallman RF. The phenomenon of reoxygenation and its implications for fractionated radiotherapy. Radiology. 1972;105:135-42. 47. Reynolds CP. Detection and treatment of minimal residual disease in high-risk neuroblastoma. Pediatric transplantation. 2004;8 Suppl 5:56-66. 48. Patel KJ, Tredan O, Tannock IF. Distribution of the anticancer drugs doxorubicin, mitoxantrone and topotecan in tumors and normal tissues. Cancer Chemother Pharmacol. 2013;72:127-38. 49. Meng F, Evans JW, Bhupathi D, Banica M, Lan L, Lorente G, et al. Molecular and cellular pharmacology of the hypoxia-activated prodrug TH-302. Molecular cancer therapeutics. 2012;11:740-51. 50. Kanno S, Higurashi A, Watanabe Y, Shouji A, Asou K, Ishikawa M. Susceptibility to cytosine arabinoside (Ara-C)-induced cytotoxicity in human leukemia cell lines. Toxicology letters. 2004;152:149-58.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 25

    Figure Legends:

    Figure 1. Increased cytotoxicity of evofosfamide under prolonged hypoxic conditions with NBL

    and RMS cells. A panel of 5 different NBL cell lines (CHLA-15, CHLA-20, CHLA-90, SK-N-

    BE(2) and SH-SY5Y) and 3 rhabdomyosarcoma cell line (RH4, RH30 and RD) were selected to

    assess the effects of evofosfamide on cell proliferation in vitro. Cell viability was measured by

    Alamar Blue assays after exposing tumor cells to increasing concentrations of evofosfamide in

    vitro for 72 hours under both normoxic and two hypoxic conditions, 1% O2 for 2 hours or 1% O2

    overnight. Dose-response curves were fit to the data and IC50 values were calculated by

    GraphPad Prism software.

    Figure 2. Anti-tumor effects of evofosfamide and topotecan in NBL xenograft models. A, A

    total of 1 × 106 cells were implanted subcutaneously into SCID/SCID mice. Once tumors were

    palpable (about 0.25cm3), mice were randomized into vehicle control (n=10) or three treatment

    groups (n = 10 for each group). Mice bearing human neuroblastoma (SK-N-BE(2) and CHLA-

    20) xenografts were treated for 2 weeks with evofosfamide (50 mg/kg; qd × 5/wk, IP) or

    topotecan (1 mg/kg; qd × 5/wk, orally) as single agents or in combination. The shaded area

    indicates the period of drug treatment. Tumor growth curves were plotted with the tumor

    volumes at different time points. One-way ANOVA was used to compare tumor volumes

    between experimental groups at different time points during the experimental period (*P <

    0.05, **P < 0.01). Data are expressed as mean ± s.e.m. After 1 cycle of therapy, the combination

    treatment with evofosfamide and topotecan led to complete tumor regression in both SK-N-

    BE(2) and CHLA-20 xenograft models. In the CHLA-20 model, tumor recurrence was observed

    in the combined treatment group. On day 55, we re-challenged the tumor bearing animals with

    the original treatment dosage and schedule on day 55 (identified by shaded area between day 55

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 26

    and 69).All the relapsed tumors remained responsive to combined treatment. B, Recurrence-free

    interval curves were estimated in CHLA-20 xenograft model by the Kaplan–Meier method and

    compared between topotecan and combination of evofosfamide and topotecan treatment using

    the log-rank (Mantel-Cox) analysis. The tumor-free interval was defined from the end of

    treatment to the first appearance of a palpable mammary tumor at least 100 mm3 in size. *, P <

    0.05. C, Mouse body weight was measured in CHLA-20 tumor-bearing mice in different

    treatment groups and control animals. Percentage of body weight loss was calculated and plotted

    to monitor the potential drug toxicity. D, Kaplan-Meier survival curves of the SK-N-BE(2)

    metastatic tumor model. NOD/SCID mice (n = 8/group) were injected with 1 × 106 SK-N-BE(2)

    cells intravenously. 14 days after tumor cell inoculation, mice were treated for 2 weeks with

    control vehicle, evofosfamide, topotecan or the combination of evofosfamide and topotecan with

    the same dose as indicated above. Statistical differences between the various groups were

    compared pair-wise using log-rank (Mantel-Cox) analysis. *, P < 0.05.

    Figure 3. Effects of evofosfamide and topotecan on the growth of rhabdomyosarcoma

    xenografts. A total of 1 × 106 cells were implanted subcutaneously into SCID/SCID mice. Once

    tumors were palpable (about 0.25cm3), mice were randomized into vehicle control group (n=10)

    or three treatment arms (n = 10 for each group). RH4 and RD xenografts were treated with

    evofosfamide (50 mg/kg; qd × 5/wk, IP) or topotecan (1 mg/kg; qd × 5/wk, orally) as single

    agents or in combination. The shaded area indicates the period of drug treatment. Tumor growth

    was monitored as described above. One-way ANOVA was used to compare tumor volumes

    between experimental groups at different time points during the experimental period (*P <

    0.05, **P < 0.01). Data are expressed as mean ± s.e.m.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 27

    Figure 4. Localization of tumor cell apoptosis, proliferation and tumor hypoxia after

    evofosfamide and topotecan treatment. RH4 xenografts from different regimens were harvested 4

    days after drug treatment for immunohistochemical analysis. Hypoxic regions were identified as

    the area of pimonidazole positive staining (circled area). A, Apoptotic cells were detected by

    anti-cleaved caspase-3 antibody. B, Ten high-power fields per treatment arm were quantified for

    cell apoptosis in both tumor hypoxic and normoxic regions expressed by percentage of positive

    cleaved caspase-3 staining area using ImageJ software. Statistical analysis was done by t test

    (*,P < 0.05; **, P < 0.01). Error bars, SD. C, Immunohistochemical staining of Ki67 was

    performed to identify proliferating cells in different groups of xenografts. D, Cell proliferation in

    normoxic regions was quantified by percentage of positive Ki67 staining area using ImageJ

    software. Statistical analysis was done by t test (*,P < 0.05; **, P < 0.01). Data are means ±

    SEM.

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • 28

    Table 1: Comparison of IC50 concentrations of evofosfamide when tested alone and in

    combination with 20nM topotecan in neuroblastoma and rhabdomyosarcoma cell lines.

    IC50 (µM) P Value Evofosfamide Evofosfamide+Topotecan

    NBL cell lines: CHLA-15-normoxia 4.6 ± 5.8 3 ± 3.5 0.39 CHLA-15-1% O2; 2hr 9.5 ± 6.3 7.4 ± 4.1 0.57 CHLA-15-1% O2; O/N 0.07 ± 0.03 0.04 ± 0.01

  • 29

    Table 2: IC50 of topotecan in neuroblastoma and rhabdomyosarcoma cell lines

    IC50 (nM) p Value1 Normoxia 1% O2, 2hr 1% O2, O/N

    NBL cell lines: CHLA-15 17.6 ± 3.9 15.6 ± 0.9 13.7 ± 1.4 NS2

    CHLA-20 12.9 ± 1.2 12.4 ± 1.0 14.1 ± 0.6 NS CHLA-90 4170 ± 1030 4550 ± 1590 5260 ± 724 NS

    SK-N-BE(2) 96.2 ± 11.8 110 ± 13.8 95.0 ± 14.3 NS SH-SY5Y 19.2 ± 7.0 12.4 ± 1.1 14.1 ± 0.6 NS

    RMS cell lines: RH4 132 ± 10.0 110 ± 21.8 119 ± 3.7 NS

    RH30 2251 ± 685 1963 ± 416 1621 ± 174 NS RD 387 ± 55.8 250 ± 87.5 317 ± 22.9 NS

    1 p value from one-way ANOVA.

    2 NS: not significant, p>0.05

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • Fig. 1

    B

    A

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • Fig. 2

    B C

    A

    D

    P

  • Fig. 3

    ** **

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • Fig. 4

    Control

    Evofosfamide

    Topotecan

    Evofosfamide + Topotecan

    DAPI Anti-cleaved

    caspase-3 Anti-

    pimonidazole Superimpose

    B

    A

    **

    *

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • Control

    Evofosfamide

    Topotecan

    Evofosfamide + Topotecan

    Anti-Ki67 Anti-pimonidazole Superimpose

    Fig. 4

    D

    C

    **

    **

    *

    *

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst December 30, 2015.Clin Cancer Res Libo Zhang, Paula Marrano, Bing Wu, et al. rhabdomyosarcoma preclinical models

    andhypoxia-activated prodrug, evofosfamide, in neuroblastoma Combined antitumor therapy with metronomic topotecan and

    Updated version

    10.1158/1078-0432.CCR-15-1853doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2016/06/02/1078-0432.CCR-15-1853.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/early/2015/12/30/1078-0432.CCR-15-1853To request permission to re-use all or part of this article, use this link

    on April 3, 2021. © 2015 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on December 30, 2015; DOI: 10.1158/1078-0432.CCR-15-1853

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-15-1853http://clincancerres.aacrjournals.org/content/suppl/2016/06/02/1078-0432.CCR-15-1853.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2015/12/30/1078-0432.CCR-15-1853http://clincancerres.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4