Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


LCL161, a SMAC mimetic, was tested against the PPTP in vitro panel (1.0 nM to 10.0 µM) and the PPTP in vivo panels (30 or 75 mg/kg [solid tumors] or 100 mg/kg [ALL]) administered orally twice in a week. LCL161 showed a median relative IC(50) value of >10 µM, being more potent against several leukemia and lymphoma lines. In vivo LCL161 induced significant differences in EFS distribution in approximately one-third of solid tumor xenografts (osteosarcoma and glioblastoma), but not in ALL xenografts. No objective tumor responses were observed. In vivo LCL161 demonstrated limited single agent activity against the pediatric preclinical models studied.

Free full text 


Logo of nihpaLink to Publisher's site
Pediatr Blood Cancer. Author manuscript; available in PMC 2013 Apr 1.
Published in final edited form as:
PMCID: PMC3253328
NIHMSID: NIHMS284344
PMID: 21681929

Initial Testing (Stage 1) of LCL161, a SMAC Mimetic, by the Pediatric Preclinical Testing Program

Abstract

LCL161, a SMAC mimetic, was tested against the PPTP in vitro panel (1.0 nM to 10.0 μM) and the PPTP in vivo panels (30 mg/kg or 75 mg/kg [solid tumors] or 100 mg/kg [ALL]) administered orally twice weekly. LCL161 showed a median relative IC50 value of >10 μM, being more potent against several leukemia and lymphoma lines. In vivo LCL161 induced significant differences in EFS distribution in approximately one-third of solid tumor xenografts (osteosarcoma, glioblastoma), but in no ALL xenografts. No objective tumor responses were observed. In vivo LCL161 demonstrated limited single agent activity against the pediatric preclinical models studied.

Keywords: Preclinical Testing, Developmental Therapeutics, SMAC mimetic

INTRODUCTION

The process of apoptosis plays a critical role in the development and homeostasis of multicellular organisms. In many cancers the cell death machinery is inhibited by the upregulation of antiapoptotic proteins, suggesting that the restoration of apoptotic activity might be an effective approach for treating these cancers. Members of the inhibitor of apoptosis (IAP) family of proteins are upregulated in many cancers, and they initially were thought to primarily contribute to oncogenesis through their ability to directly inhibit members of the caspase family of apoptotic enzymes. It now appears that only one member of the family (XIAP) acts as a direct inhibitor of caspases [1], and that it is able to inhibit the enzymatic activity of both initiator (caspase-9) and executor (caspase-3 and -7) caspases. A more recently recognized oncogenic role for IAP family members is in regulating NF-κB signaling [2].

The second mitochondria-derived activator of caspases (Smac) has a unique function in regulating apoptosis. In non-stressed cells Smac is sequestered in mitochondria and is released into the cytosol only upon induction of mitochondrial dysfunction or apoptosis [3,4]. Cytosolic Smac selectively binds to IAPs through conserved Baculovirus IAP Repeat (BIR) domains [5,6], promoting cell death. Consequently, small molecule drugs that mimic the interaction of Smac with IAPs (Smac mimetics) have been designed. Smac mimetics bind with high affinity to IAPs, including XIAP, cIAP1 and cIAP2. Surprisingly, cell death induced by Smac mimetics was found to require TNFα signaling and caspase-8 and to be independent of caspase-9 [7]. Work from a number of laboratories has shown that Smac mimetics rapidly induce auto-ubiquitylation and proteasomal degradation of cIAP1 and cIAP2 resulting in the activation of non-canonical NF-κB signaling and subsequent increased TNFα production and autocrine stimulation of TNFR1 [710]. This increased TNFα signaling leads to caspase-8 activation and apoptosis as a result of the enhanced RIPK1 levels that are a downstream effect of reduced cIAP ubiquitylation of RIPK1 [810].

LCL161 is a small molecule drug mimetic of Smac that binds to IAPs with high affinity and initiates the destruction of cIAP1 and cIAP2 [11]. LCL161 induces apoptosis in some cancer cell lines and potentiates the effects of tyrosine kinase inhibition against leukemic disease [11,12]. LCL161 is currently in clinical trials as a single agent or in combination with cytotoxic agents [13].

MATERIALS AND METHODS

In vitro testing

In vitro testing was performed using DIMSCAN, as previously described [14]. Cells were incubated in the presence of LCL161 for 96 hours at concentrations from 1 nM to 10 μM and analyzed as previously described [15].

In vivo tumor growth inhibition studies

CB17SC scid−/− female mice (Taconic Farms, Germantown NY), were used to propagate subcutaneously implanted kidney/rhabdoid tumors, sarcomas (Ewing, osteosarcoma, rhabdomyosarcoma), neuroblastoma, and non-glioblastoma brain tumors, while BALB/c nu/nu mice were used for glioma models, as previously described [16,17]. Human leukemia cells were propagated by intravenous inoculation in female non-obese diabetic (NOD)/scid−/− mice as described previously [18]. Female mice were used irrespective of the patient gender from which the original tumor was derived. All mice were maintained under barrier conditions and experiments were conducted using protocols and conditions approved by the institutional animal care and use committee of the appropriate consortium member. Eight to ten mice were used in each control or treatment group. Tumor volumes (cm3) [solid tumor xenografts] or percentages of human CD45-positive [hCD45] cells [ALL xenografts] were determined as previously described [16]. Responses were determined using three activity measures as previously described [16]. An in-depth description of the analysis methods is included in the Supplemental Response Definitions section.

Statistical Methods

The exact log-rank test, as implemented using Proc StatXact for SAS®, was used to compare event-free survival distributions between treatment and control groups. P-values were two-sided and were not adjusted for multiple comparisons given the exploratory nature of the studies.

Drugs and Formulation

LCL161 was provided to the Pediatric Preclinical Testing Program by Novartis Pharmaceuticals, through the Cancer Therapy Evaluation Program (NCI). LCL161 was tested initially using a 30 mg/kg dose administered by oral gavage twice weekly (Mon-Tues) repeated weekly for a planned treatment duration of 6 weeks. Subsequently, limited testing at 75 mg/kg (solid tumors) or 100 mg/kg (ALL models) was undertaken using the same schedule and route of drug administration. LCL161 was formulated for oral gavage by dissolving in 0.1N HCl, and diluting to volume with sodium acetate buffer (100 mM, pH 4.63) to produce a solution with pH 4.3–4.6. LCL161 was provided to each consortium investigator in coded vials for blinded testing.

RESULTS

In vitro testing

LCL161 was evaluated against the the 23 cell lines in the PPTP in vitro panel using 96 hour exposure to concentrations ranging from 1.0 nM to 10.0 μM. LCL161 achieved 50% growth inhibition (i.e., Ymin < 50%) against only 3 of the 23 tested PPTP cell lines, Table I. The three cell lines included two T-cell ALL cell lines (COG-LL-317 and CCRF-CEM) and an anaplastic large cell lymphoma cell line (Karpas-299), with CCRF-CEM and Karpas-299 showing the lowest relative IC50 values (0.25 and 1.6 μM, respectively).

Table I

Summary of LCL161 activity In vitro

Cell LineHistologyRelative IC50 (μM)Ymin % (Observed)
RDRhabdomyosarcoma< 1078.1
Rh41Rhabdomyosarcoma< 1058.9
Rh18Rhabdomyosarcoma< 1061.1
Rh30Rhabdomyosarcoma< 1071.6
BT-12Rhabdoid< 1086.3
CHLA-266Rhabdoid< 1068.7
TC-71Ewing sarcoma< 1084.0
CHLA-9Ewing sarcoma< 1084.4
CHLA-10Ewing sarcoma< 1064.7
CHLA-258Ewing sarcoma< 1053.3
GBM2Glioblastoma< 1072.4
NB-1643Neuroblastoma< 1094.1
NB-EBc1Neuroblastoma< 1080.5
CHLA-90Neuroblastoma< 1073.6
CHLA-136Neuroblastoma< 1097.0
NALM-6ALL< 1050.6
COG-LL-317ALL9.346.8
RS4;11ALL< 1063.9
MOLT-4ALL< 1075.3
CCRF-CEMALL0.2512.5
Kasumi-1AML< 1053.4
Karpas-299ALCL1.604.9
Ramos-RA1NHL< 1052.2
Median < 10 68.7
Minimum 0.25 4.9
Maximum < 10 97.0

ALCL, anaplastic large cell lymphoma.

In vivo testing

LCL161 was tested using a 30 mg/kg dose administered by oral gavage twice weekly (Mon-Tues) repeated weekly for a planned treatment duration of 6 weeks. The dose was reduced below the planned 100 mg/kg dose because of the results of toxicity testing in SCID mice. However, toxicity in tumored mice was similar in control and treatment groups (1.6%). All 46 tested xenograft models were considered evaluable for efficacy. Complete details of testing are provided in Supplemental Tables I and II, including total numbers of mice, number of mice that died (or were otherwise excluded), numbers of mice with events and average times to event, tumor growth delay, as well as numbers of responses and T/C values.

LCL161 induced significant differences in EFS distribution compared to controls in 12 of 38 evaluable solid tumor xenografts (32%) tested. Significant tumor growth delay was observed in multiple solid tumor panels, but was most consistently present in the osteosarcoma (5 of 6), glioblastoma (2 of 4) and neuroblastoma (2 of 6) panels. None of the 8 evaluable ALL xenografts showed a significant difference in EFS distribution between treated and control animals.

Criteria for intermediate activity for the time to event activity measure (i.e., EFS T/C > 2) were met in 1 of 36 (3%) solid tumor xenografts evaluable for this measure (Table II). Intermediate activity was observed for the medulloblastoma xenograft BT-28. No ALL xenografts met criteria for intermediate activity for the EFS T/C activity measure. Using the PPTP Objective Response Measure, objective response (i.e., tumor regression) was scored for a single xenograft, BT-28 (medulloblastoma). However, responses in individual animals were widely divergent for this xenograft, with 5 tumors regressing completely and four tumors progressing (PD1), giving a median `score' consistent with PR. Among the remaining solid tumor xenografts, 5 showed a PD2 response (progressive disease with growth delay). PD2 responses occurred across multiple panels. There were no objective responses (PR or CR) among the ALL xenografts.

Table II

Summary of LCL161 Activity In Vivo

Xenograft LineHistologyMedian Time to EventP-valueEFS T/CMedian Final RTVT/CP-valueT/C ActivityEFS ActivityResponse Activity
30 mg/kg
BT-29Rhabdoid12.50.3791.0>40.940.481LowLowLow
KT-14Rhabdoid31.70.5781.0>41.040.684LowLowLow
KT-12Rhabdoid10.00.4651.1>40.790.353LowLowLow
KT-10Wilms14.30.7010.9>41.090.796LowLowLow
KT-11Wilms11.50.5181.0>41.160.631LowLowLow
KT-13Wilms10.90.0931.1>41.070.796LowLowLow
SK-NEP-1Ewing9.00.3101.0>40.810.143LowLowLow
EW5Ewing14.4<0.0011.5>40.740.009LowLowInt
EW8Ewing11.70.0731.8>40.460.165LowLowInt
TC-71Ewing9.70.9711.0>41.230.853LowLowLow
CHLA258Ewing8.30.7581.2>41.180.971LowLowLow
Rh10ALV RMS25.10.9321.3>40.950.965LowLowLow
Rh28ALV RMS13.10.3420.7>41.430.052LowLowLow
Rh30ALV RMS15.30.1861.4>40.800.315LowLowLow
Rh30RALV RMS12.20.2711.2>40.660.002LowLowLow
Rh41ALV RMS15.40.5861.0>41.001.000LowLowLow
Rh18EMB RMS13.5<0.0011.2>40.630.002LowLowLow
BT-28Medulloblastoma> EP0.002>10.9>40.420.009IntIntHigh
BT-45Medulloblastoma21.30.0511.3>40.660.165LowLowLow
BT-50Medulloblastoma> EP0.204> 1.32.80.790.014LowNEInt
BT-41Ependymoma> EP1.000.2.00.570.123LowNEInt
BT-44Ependymoma9.60.7070.9>41.310.842LowLowLow
GBM2Glioblastoma9.70.7090.9>40.970.009LowLowLow
BT-39Glioblastoma11.90.0031.8>40.650.123LowLowInt
D645Glioblastoma9.70.1200.8>41.190.165LowLowLow
D456Glioblastoma11.80.0451.5>40.820.481LowLowLow
NB-SDNeuroblastoma14.90.7141.4>40.830.008LowLowLow
NB-1771Neuroblastoma23.40.0081.4>40.520.853LowLowLow
NB-1691Neuroblastoma10.60.4541.1>40.900.696LowLowLow
NB-EBc1Neuroblastoma14.80.0241.4>40.760.234LowLowLow
CHLA-79Neuroblastoma18.50.1461.5>40.800.579LowLowLow
NB-1643Neuroblastoma8.60.7481.0>41.040.035LowLowLow
OS-1Osteosarcoma38.80.0171.3>40.760.001LowLowLow
OS-2Osteosarcoma23.6<0.0011.3>40.760.278LowLowLow
OS-17Osteosarcoma20.80.5001.0>40.91<0.001LowLowLow
OS-9Osteosarcoma28.6<0.0011.8>40.57<0.001LowLowInt
OS-33Osteosarcoma26.1<0.0011.4>40.610.133LowLowLow
OS-31Osteosarcoma23.50.0441.1>40.81LowLowLow
ALL-2ALL B-precursor10.40.5311.0>25.LowLow
ALL-3ALL B-precursor9.30.3631.2>25.LowLow
ALL-4ALL B-precursor9.00.8191.5>25.LowLow
ALL-7ALL B-precursor6.01.0001.0>25.LowLow
ALL-8ALL T-cell11.20.5471.0>25.LowLow
ALL-16ALL T-cell> EP0.103> 1.4>25.NEInt
ALL-17ALL B-precursor6.30.6691.1>25.LowLow
ALL-19ALL B-precursor5.40.6521.1>25.LowLow
75 mg/kg (solid Tumors)
EW5Ewing11.5<0.0011.7>40.68<0.001LowLowLow
BT-28Medulloblastoma5.70.31901.2>40.660'165LowLowLow
BT-39Glioblastoma18.10.00601.3>40.66LowLowLow
Karpas-299ALCL13.3<0.0011.6>40.65LowLowLow
100 mg/kg (ALL models)
ALL-7ALL B-precursor3.00.68801.0>25.LowLow
ALL-19ALL B-precursor10.20.52402.1>25.LowLow
ALL-31ALL T-cell5.80.48901.0>25.LowLow
MLL-7ALL B-precursor8.90.21501.7>25.LowLow

Because of the limited toxicity observed in the initial testing at 30 mg/kg, LCL161 was re-tested at higher doses against selected solid tumor models (75 mg/kg) and ALL models (100 mg/kg) using the same schedule of drug administration. LCL161 was well tolerated at this higher dose (1 of 60 deaths in treatment groups). Against EW-5 and BT-39 glioblastoma LCL161 significantly inhibited growth, whereas against BT-28 tumors LCL161 did not significantly inhibit growth (PD1 responses), and hence did not confirm the PR at the lower dose level. LCL161 was ineffective against ALL 7 (B-precursor), ALL 19 (T-cell), ALL 31 (T-cell) and MLL 7 (B-precursor) leukemia models (Table II). The anaplastic large cell lymphoma line, Karpas-299, was the most sensitive cell line in the in vitro panel, and hence we tested its sensitivity (75 mg/kg dose level) in vivo maintained as a subcutaneous xenograft. LCL161 significantly inhibited growth of the Karpas-299 xenograft with an EFS T/C value of 1.6, but tumor regression was not observed (Table II).

DISCUSSION

The limited level of in vitro activity observed for LCL161 by the PPTP is consistent with results for adult cancer cell lines showing that LCL161 demonstrates activity against a minority of cell lines [11]. However, while some adult cancer cell lines have IC50 values to LCL161 in the 20 to 50 nM range, there are no pediatric cell lines in the PPTP panel that show this degree of sensitivity. A report describing the activity of another small molecule SMAC-mimetic against 50 non-small cell lung cancer (NSCLC) cell lines also showed activity against a minority (approximately 15%) of cell lines [19]. Similarly, a SMAC-mimetic developed by Abbott Laboratories showed activity against approximately 15% of 59 cell lines studied [20]. The three PPTP cell lines showing the greatest sensitivity to LCL161 were all lymphoid derived: CCRF-CEM (T-cell ALL) and Karpas-299 (ALCL) and COG-LL-317 (T-cell ALL). In vivo, LCL161 had limited activity causing growth delay in a subset of PPTP xenografts, with a single tumor line BT-28 meeting the criteria for PR at 30 mg/kg, but that was not reproducible at 75 mg/kg. There were no objective responses in the leukemia models at either dose level evaluated. Thus, LCL161 as a single agent demonstrated a low level of activity in this screen.

An evaluation of LCL161 activity against adult cancer cell lines demonstrated that canonical and non-canonical NFκB signaling are not differentially activated in sensitive and resistant cells, but that TNFα is induced only in the former [11]. Furthermore, high baseline TNFα levels appeared to predispose to sensitivity. We therefore examined TNFα expression levels for PPTP cell lines and xenografts generated using Affymetrix U133 Plus 2.0 arrays [21] (Supplemental Figure 1). Few cell lines or xenografts showed elevated TNFα expression. One of the two most sensitive cell lines (Karpas-299) showed elevated TNFα expression, as did a glioblastoma xenograft (BT-39) that showed significant growth delay to LCL161. Multiple B-precursor ALL xenografts showed moderately elevated TNFα expression, but LCL161 did not show significant in vivo activity against these xenografts.

In summary, LCL161 showed limited in vitro and in vivo activity as a single agent against the PPTP's childhood cancer preclinical models. Future work evaluating small molecule Smac mimetics such as LCL161 in the childhood cancer setting can focus on their utility in combination with standard cytotoxic agents, signaling pathway inhibitors [12], and activators of the extrinsic cell death pathway such as TRAIL [22,23].

Supplementary Material

Supp Figure S1

Supp Table S1

Supp Table S2

Supplementary Data

ACKNOWLEDGEMENTS

This work was supported by NO1-CM-42216, CA21765, and CA108786 from the National Cancer Institute, and LCL161 was provided by Novartis Pharmaceuticals. In addition to the authors represents work contributed by the following: Sherry Ansher, Joshua Courtright, Edward Favours, Henry S. Friedman, Debbie Payne-Turner, Charles Stopford, Chandra Tucker, Amy Wozniak, Joe Zeidner, Ellen Zhang, and Jian Zhang. Children's Cancer Institute Australia for Medical Research is affiliated with the University of New South Wales and Sydney Children's Hospital.

Footnotes

CONFLICT OF INTEREST STATEMENT: The authors consider that there are no actual or perceived conflicts of interest.

Reference List

1. Eckelman BP, Salvesen GS, Scott FL. Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep. 2006;7(10):988–994. [Europe PMC free article] [Abstract] [Google Scholar]
2. Gyrd-Hansen M, Meier P. IAPs: from caspase inhibitors to modulators of NF-kappaB, inflammation and cancer. Nat Rev Cancer. 2010;10(8):561–574. [Abstract] [Google Scholar]
3. Chai J, Du C, Wu JW, et al. Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature. 2000;406(6798):855–862. [Abstract] [Google Scholar]
4. Du C, Fang M, Li Y, et al. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell. 2000;102(1):33–42. [Abstract] [Google Scholar]
5. Liu Z, Sun C, Olejniczak ET, et al. Structural basis for binding of Smac/DIABLO to the XIAP BIR3 domain. Nature. 2000;408(6815):1004–1008. [Abstract] [Google Scholar]
6. Srinivasula SM, Hegde R, Saleh A, et al. A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature. 2001;410(6824):112–116. [Abstract] [Google Scholar]
7. Gaither A, Porter D, Yao Y, et al. A Smac mimetic rescue screen reveals roles for inhibitor of apoptosis proteins in tumor necrosis factor-alpha signaling. Cancer Res. 2007;67(24):11493–11498. [Abstract] [Google Scholar]
8. Varfolomeev E, Goncharov T, Fedorova AV, et al. c-IAP1 and c-IAP2 are critical mediators of tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB activation. J Biol Chem. 2008;283(36):24295–24299. [Europe PMC free article] [Abstract] [Google Scholar]
9. Mahoney DJ, Cheung HH, Mrad RL, et al. Both cIAP1 and cIAP2 regulate TNFalpha-mediated NF-kappaB activation. Proc Natl Acad Sci U S A. 2008;105(33):11778–11783. [Europe PMC free article] [Abstract] [Google Scholar]
10. Bertrand MJ, Milutinovic S, Dickson KM, et al. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol Cell. 2008;30(6):689–700. [Abstract] [Google Scholar]
11. Zawel LS, Straub C, Firestone B, et al. Therapeutic targeting of inhibitor of apoptosis proteins. 101st Annual Meeting of the American Association for Cancer Research; 2010. Abstr #138. [Google Scholar]
12. Weisberg E, Ray A, Barrett R, et al. Smac mimetics: implications for enhancement of targeted therapies in leukemia. Leukemia. 2010;24(12):2100–2109. [Europe PMC free article] [Abstract] [Google Scholar]
13. Infante JR, Dees EC, Burris HA, et al. A phase I study of LCL161, an oral IAP inhibitor, in patients with advanced cancer. 101st Annual Meeting of the American Association for Cancer Research; 2010. Abstr #2775. [Google Scholar]
14. Frgala T, Kalous O, Proffitt RT, et al. A fluorescence microplate cytotoxicity assay with a 4-log dynamic range that identifies synergistic drug combinations. Mol Cancer Ther. 2007;6(3):886–897. [Abstract] [Google Scholar]
15. Houghton PJ, Morton CL, Kolb EA, et al. Initial testing (stage 1) of the proteasome inhibitor bortezomib by the pediatric preclinical testing program. Pediatr Blood Cancer. 2007 [Abstract] [Google Scholar]
16. Houghton PJ, Morton CL, Tucker C, et al. The pediatric preclinical testing program: Description of models and early testing results. Pediatr Blood Cancer. 2006 [Abstract] [Google Scholar]
17. Keir ST, Morton CL, Billups C, et al. Initial testing of VNP40101M (Cloretazine) by the pediatric preclinical testing program. Pediatr Blood Cancer. 2008;51(3):439–441. [Europe PMC free article] [Abstract] [Google Scholar]
18. Liem NL, Papa RA, Milross CG, et al. Characterization of childhood acute lymphoblastic leukemia xenograft models for the preclinical evaluation of new therapies. Blood. 2004;103(10):3905–3914. [Abstract] [Google Scholar]
19. Oost TK, Sun C, Armstrong RC, et al. Discovery of potent antagonists of the antiapoptotic protein XIAP for the treatment of cancer. J Med Chem. 2004;47(18):4417–4426. [Abstract] [Google Scholar]
20. Petersen SL, Wang L, Yalcin-Chin A, et al. Autocrine TNFalpha signaling renders human cancer cells susceptible to Smac-mimetic-induced apoptosis. Cancer Cell. 2007;12(5):445–456. [Europe PMC free article] [Abstract] [Google Scholar]
21. Neale G, Su X, Morton CL, et al. Molecular characterization of the pediatric preclinical testing panel. Clin Cancer Res. 2008;14(14):4572–4583. [Europe PMC free article] [Abstract] [Google Scholar]
22. Li L, Thomas RM, Suzuki H, et al. A small molecule Smac mimic potentiates TRAIL- and TNFalpha-mediated cell death. Science. 2004;305(5689):1471–1474. [Abstract] [Google Scholar]
23. Fulda S, Wick W, Weller M, et al. Smac agonists sensitize for Apo2L/TRAIL- or anticancer drug-induced apoptosis and induce regression of malignant glioma in vivo. Nat Med. 2002;8(8):808–815. [Abstract] [Google Scholar]

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/5580858
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/5580858

Article citations


Go to all (52) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


Funding 


Funders who supported this work.

NCI NIH HHS (7)

National Cancer Institute (3)