Purpose: Enhanced tumor cell survival through expression of inhibitors of apoptosis (IAP) is a hallmark of cancer. Survivin, an IAP absent from most normal tissues, is overexpressed in many malignancies and associated with a poorer prognosis. We report the first-in-human dose study of LY2181308, a second-generation antisense oligonucleotide (ASO) directed against survivin mRNA.

Patients and Methods: A dose-escalation study evaluating the safety, pharmacokinetics, and pharmacodynamics of LY2181308 administered intravenously for 3 hours as a loading dose on 3 consecutive days and followed by weekly maintenance doses. Patients were eligible after signing informed consent, had exhausted approved anticancer therapies and agreed to undergo pre- and posttreatment tumor biopsies to evaluate reduction of survivin protein and gene expression.

Results: A total of 40 patients were treated with LY2181308 at doses of 100 to 1,000 mg. Twenty-six patients were evaluated at the recommended phase 2 dose of 750 mg, at which level serial tumor sampling and [11C]LY2183108 PET (positron emission tomography) imaging demonstrated that ASO accumulated within tumor tissue, reduced survivin gene and protein expression by 20% and restored apoptotic signaling in tumor cells in vivo. Pharmacokinetics were consistent with preclinical modeling, exhibiting rapid tissue distribution, and terminal half-life of 31 days.

Conclusions: The tumor-specific, molecularly targeted effects demonstrated by this ASO in man underpin confirmatory studies evaluating its therapeutic efficacy in cancer.

Translational Relevance

Evasion of apoptosis is a hallmark of malignancy. Survivin is a small, naturally occurring inhibitor of apoptosis (IAP) that is often highly expressed by cancer cells making it an attractive target for therapeutic intervention. The antisense oligonucleotide (ASO) LY2181308 downregulates survivin by targeting the survivin transcript and has antitumor efficacy through induction of tumor cell apoptosis in preclinical models. This First-In-Human (FHD) study demonstrates the proof of concept that ASO therapy directed against survivin mRNA reduces survivin mRNA and protein levels and restores apoptosis in tumors of cancer patients. The research represents a critical step in the translation from discovery of survivin, an important tumor-related IAP, to successful clinical application of survivin-targeted therapy. Finally, this FHD study helps to better understand how ASOs can be used as novel agents in the treatment of cancer.

Survivin, a 16.5-kDa protein encoded by the essential gene BIRC5, was originally identified as an inhibitor of apoptosis (IAP) that exerts its effects through binding to SMAC (second mitochondrial activator of caspases) preventing the sequestration of IAPs by SMAC and inhibition of caspase-dependent apoptosis (1). As a component of the kinetochore-associated complex, survivin also plays an important role in the regulation of late mitosis and cytokinesis (2). Survivin is expressed in a wide range of human cancers and when overexpressed, is associated with a poorer prognosis (3). With the exception of placenta, thymus, activated T cells, gastrointestinal crypt cells, and regenerating liver, survivin is not expressed in normal adult tissue (4). Thus, survivin represents an attractive molecular target for therapeutic intervention. Targeted approaches against survivin include small molecule inhibitors against the survivin protein, gene silencing, and survivin mRNA blockade (5, 6). The 2′-O-methoxyethyl–modified ASO (second generation) LY2181308 is an 18-mer ASO that binds to the translation initiation codon of the survivin transcript. Following ASO hybridization to survivin mRNA, RNase H–dependent cleavage of the duplex ensues with subsequent degradation of the survivin mRNA (7). Thus, survivin protein expression is specifically inhibited without affecting expression of other genes including other IAP proteins (8). Compared with phosphorothioate or first-generation ASOs, second-generation ASOs are more stable, have an improved pharmacokinetic (PK) profile, increased potency, and reduced toxicity (9). Using aggregate data from preclinical pharmacology and toxicology, an integrated PK/PD (pharmacodynamic) model was developed to predict a biologically effective dose range for clinical evaluation (10). On the basis of this, a first-in-human dose (FHD) study was designed to evaluate the biodistribution profile of LY2181308 by measuring the following tumor-specific PD changes: tumor tissue penetration of LY2181308, downregulation of tumor survivin at the mRNA, and protein levels and restoration of tumor apoptosis at a dose and schedule that was safe in humans. To this end, pre- and posttreatment tumor sampling was performed for rigorous assessment of target modulation (i.e., survivin protein and gene expression), the effect of restoring normal apoptosis/cell cycle–related protein activity and biodistribution of LY2181308 including subcellular localization.

Study design

This FHD monotherapy study was divided into 3 parts: part A (safety, PK), 1-patient cohorts with initial 100 mg and escalated, by dose doubling, to 400 mg; part B (safety, PK, PD), 3-patient cohorts with 50% dose escalation until dose-limiting toxicity (DLT); part C (dose confirmation cohort, PK, PD). The transition from part A to part B was planned on the basis of either development of toxicity or evidence from PK that the predicted biological effective dose (BED) had been reached. The recommended dose for phase 2 studies was determined from both DLT and the reduction in survivin expression in tumor tissue at the anticipated BED as defined by the preclinical PK/PD model of Callies et al. (10). At the recommended phase 2 dose, 2 companion studies were conducted at the Universities of Oxford (study 1, endobronchial tumor sampling) and Manchester (study 2, [11C]LY2181308 study). The entire study was approved by the Medicines and Healthcare products Regulatory Agency (MHRA) and a Multicentre Research Ethics Committee (MREC).

Enrollment criteria

Inclusion criteria were as follows: at least 18 years of age, confirmed malignancy, exhausted approved standard therapies, tumor accessible for biopsy, written informed consent, Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, discontinued previous anticancer therapies, absolute neutrophil count ≥ 1.5 × 109/L, platelets ≥ 100 × 109/L, hemoglobin ≥ 9 g/dL, normal serum bilirubin <2.5× upper limit of normal, alanine transaminase (ALT) and aspartate transaminase (AST), calculated creatinine clearance ≥ 50 mL/min, normal activated prothrombin time (aPTT), and prothrombin time (PT), and contraceptive precautions taken. Exclusion criteria were as follows: bleeding diathesis, major surgery within last 4 weeks, pregnant or lactating, symptomatic central nervous system (CNS) neoplasm, taking concomitant anticoagulant therapy, received prior ASO, treatment with an unapproved drug, positive test results for viral infection.

Drug formulation and schedule of administration

LY2181308 was diluted in 500 mL of normal saline for intravenous injection and infused as a 3-hour infusion on 3 consecutive days as a loading dose, followed by weekly maintenance doses. The dose and dose schedule were based on preclinical safety, PK and pharmacology studies of LY2181308. These were integrated within a PK/PD model that predicted the BED (10).

Treatment assessment

DLT was defined (CTCAE Version 3.0; ref. 11) as 1 of 3 patients with the following: grade 3/4 hematologic toxicities more than 5 days, aPTT prolongation more than 48 hours after infusion, any grade 3/4 nonhematologic toxicity or any other DLT including those associated with sustained complement activation. Laboratory examinations: C-reactive protein (CRP), complement split products (National Jewish Medical and Research Center), hematology, serum chemistry. Pretreatment tumor biopsy: after enrollment and before first loading. Posttreatment tumor biopsy: window of 48 to 96 hours after the third loading dose. Descriptive statistics were used for all patients receiving at least 1 dose of LY2181308 to evaluate safety, PK, and efficacy.

Pharmacokinetic assessment

PK parameters were analyzed (WinNonlin Enterprise, Version 5.2) for: time of maximum concentration (Tmax), maximum plasma concentration (Cmax), area under the plasma concentration versus time curve (AUC), and clearance (CL) following the third and fifth administration of LY2181308 on days 3 and 15. Plasma PK data were pooled for nonlinear mixed affect modeling analysis (NONMEM, Version 6.1) to determine the compartmental PK parameters: mean value, variance, and subject variability. The interindividual variability was coded as an exponential model and the residual variability as a proportional model.

Development of a specific antibody to detect the ASO

A polyclonal rabbit antibody against the ASO was generated using a keyhole limpet hemocyanin (KLH)–modified ASO (Lampire Biological Laboratories). It was validated for use in formalin-fixed, paraffin-embedded specimen utilizing Ventana's DiscoveryXTstaining platforms (Ventana).

Pharmacodynamic assessment

Formalin-fixed tissue was used for immunohistochemical assessment at Ventana (Tucson) to determine: ASO, survivin (NB500-201; Novus Biologicals), cleaved caspase 3 (CC3; CST9661; Cell Signaling Technologies), and Ki67 (mouse monoclonal antibody; Ventana). All antibodies were detected by appropriate secondary antibodies (Vector Laboratories). Staining intensity was determined using the HSCORE and Ventana's automated imaging software (12). Gene expression analysis (Panomics; ref. 13) was used to quantify Survivin mRNA expression (branched DNA, bDNA). Percentage change in protein and mRNA expression was summarized using medians and approximately 95% confidence limits (14).

Endobronchial biopsy

Fiberoptic endobronchial tumor sampling is a safe, short, and standard procedure (15). Endobronchial tumors were visualized and sampled by brushing with a 1-mm brush. The samples were processed immediately for flow cytometry assessment.

Flow cytometry

Briefly, disaggregated tumor cells were fixed in formaldehyde and subsequently stained with phycoerytherin-(PE)conjugated monoclonal antibodies directed against survivin (Clone 91630; R&D Systems). Flow cytometric data (obtained on a CyAN machine, Dako) were analyzed using Summit software (Dako).

[11C]LY2181308 PET imaging

After establishing safety in nonhuman primates (16), patients received less than 1 mg and less than 600 MBq of [11C]LY2181308 prior to LY2181308, and during the maintenance infusion on day 15 (17). PET data were collected for 90 minutes following bolus radiotracer injection.

FDG-PET imaging

FDG-PET imaging was performed prior to treatment with LY2181308 and on day 22. The imaging was performed as previously recommended (18) and consistent with institutional radiation guidelines.

Between October 2004 and December 2008, 40 patients were enrolled in this monotherapy FHD study, of whom 17 were enrolled in the initial dose-escalation stage with doses ranging from 100 mg to 1,000 mg. Twenty-six patients were treated at the 750 mg dose level, including 6 patients in 2 site-specific studies: study 1 assessed apoptosis and cell-cycle progression changes by flow cytometry of endobronchial tumor cells in 3 patients with NSCLC (non–small cell lung carcinoma); study 2 examined the biodistribution of [11C]LY2181308 measured by PET in 3 patients. The demographics of the study population was typical of phase 1 oncology trials (19) with the majority of patients having received prior chemotherapy (Table 1). Fourteen patients with hepatic metastasis were included, of whom 11 had biopsies taken from liver lesions. LY2181308 was well tolerated with the majority of the patients showing grade 1 or 2 toxicities (29/40, 72.5%). Of the 40 patients, 11 patients (27.5%) had grade 3 or 4 toxicities (Table 2). Two of the 11 patients received the 1,000 mg dose. One patient had a grade 3 hypophosphatemia, whereas the other patient experienced a grade 4 lymphopenia and a prolonged Grade 3 headache, which did not respond to pain medication. This event was defined as the DLT for this study. Concurrently, a sharp rise in CRP was observed in this patient raising the concern of a possible complement-induced cerebrospinal leak syndrome. Three patients were treated at the 900 mg dose level and no DLTs were observed, but CRP, AST/ALT, and moderate flu-like symptoms occured during the loading doses, suggesting continued complement-activation. The most common symptoms were flu-like syndrome (fever, rigor, musculoskeletal pain, nausea), fatigue, and vomiting (Table 2). The most frequent laboratory toxicity was prolongation of aPTT (generally grade 1), which was observed at the time of the infusion in 75% of the patients. Other laboratory abnormalities, not considered as severe adverse events by investigators, included lymphopenia (70%), thrombocytopenia (38%), hypokalemia (38%), and anemia (35%). Transient increases in complement Bb were noted at the end of the third loading dose, but was not associated with adverse clinical events. Given the comparable PK profile of the higher doses of 900 and 1,000 mg to the 750-mg dose, the recommended dose of LY2181308 for further clinical studies using the same dosing schedule was defined as 750 mg, a dose likely to be safe in combination with chemotherapeutics.

Table 1.

Baseline patient and disease characteristics (n = 40)

Sexn (%)
 Male 19 (47.5) 
 Female 21 (52.5) 
Age group  
 <65 32 (80.0) 
 >65 8 (20.0) 
ECOG performance status  
 0 15 (37.5) 
 1 25 (62.5) 
Pathologic diagnosis (n)  
 Gastrointestinal tumors (including 7 colon, 1 rectal, 2 gastric, 1 esophageal, 1 pancreas cancer) 12 (30.0) 
 Breast cancer 8 (20.0) 
 Melanoma 7 (17.5) 
 Lung cancer 7 (17.5) 
 Other (3 sarcoma, 1 ovary, 1 head and neck, 1 unknown adenocarcinoma) 6 (15.0) 
Prior therapy  
 Radiotherapy 23 (57.5) 
 Surgery 30 (75.0) 
 Chemotherapy 39 (97.5) 
Sexn (%)
 Male 19 (47.5) 
 Female 21 (52.5) 
Age group  
 <65 32 (80.0) 
 >65 8 (20.0) 
ECOG performance status  
 0 15 (37.5) 
 1 25 (62.5) 
Pathologic diagnosis (n)  
 Gastrointestinal tumors (including 7 colon, 1 rectal, 2 gastric, 1 esophageal, 1 pancreas cancer) 12 (30.0) 
 Breast cancer 8 (20.0) 
 Melanoma 7 (17.5) 
 Lung cancer 7 (17.5) 
 Other (3 sarcoma, 1 ovary, 1 head and neck, 1 unknown adenocarcinoma) 6 (15.0) 
Prior therapy  
 Radiotherapy 23 (57.5) 
 Surgery 30 (75.0) 
 Chemotherapy 39 (97.5) 
Table 2.

Study drug related adverse events occurring in 2 or more patients (n = 40)

CTCAE descriptionMaximum CTC grade
1234
Laboratory     
 Partial thromboplastin time 28   
 Platelet counts   
 ALT/SGPT (serum glutamic pyruvic transaminase)   
 Metabolic/laboratory—other (including CRP)   
 Hemoglobin   
 Lymphopenia 
 AST/SGOT (serum glutamic oxaloacetic transaminase)   
 Phosphate (hypophosphatemia)   
 Potassium (hypokalemia)    
 Glucose (hyperglycemia)   
 Leukocytes (total WBC)    
 Blood/bone marrow—other (eosinophils, etc)   
 Alkaline phosphatase   
 Bilirubin (hyperbilirubinemia)    
 Sodium (hyponatremia)   
Nonlaboratory     
 Fever (in the absence of neutropenia)  
 Nausea  
 Fatigue (asthenia, lethargy, malaise)  
 Vomiting   
 Diarrhea   
 Pain neurology—head/headache   
 Rigors/chills   
 Flu-like syndrome   
 Hypotension   
 Pain musculoskeletal–joint    
 Sweating (diaphoresis)    
CTCAE descriptionMaximum CTC grade
1234
Laboratory     
 Partial thromboplastin time 28   
 Platelet counts   
 ALT/SGPT (serum glutamic pyruvic transaminase)   
 Metabolic/laboratory—other (including CRP)   
 Hemoglobin   
 Lymphopenia 
 AST/SGOT (serum glutamic oxaloacetic transaminase)   
 Phosphate (hypophosphatemia)   
 Potassium (hypokalemia)    
 Glucose (hyperglycemia)   
 Leukocytes (total WBC)    
 Blood/bone marrow—other (eosinophils, etc)   
 Alkaline phosphatase   
 Bilirubin (hyperbilirubinemia)    
 Sodium (hyponatremia)   
Nonlaboratory     
 Fever (in the absence of neutropenia)  
 Nausea  
 Fatigue (asthenia, lethargy, malaise)  
 Vomiting   
 Diarrhea   
 Pain neurology—head/headache   
 Rigors/chills   
 Flu-like syndrome   
 Hypotension   
 Pain musculoskeletal–joint    
 Sweating (diaphoresis)    

Abbreviation: CTC, circulating tumor cells.

The PK properties of LY2181308 and reduction of survivin expression in tumor tissue were assessed by collecting plasma samples and tumor biopsies before and after completing the loading dose. In parts B and C of the protocol, 31 patients were enrolled with 26 (84%) agreeing to have both pre- and posttreatment biopsies. Ten patients had either insufficient tumor material in the posttreatment biopsy or endogenous pigments (bilirubin, hemosiderin, and melanin) that interfered with IHC staining. Hence, in 16 (52%) patients, we obtained sufficient pre- and postdosing tumor tissue to assess the PD effects of LY2181308 by protocol-defined laboratory procedures (Fig. 1A–F).

Fig. 1.

Reduction of survivin protein and mRNA expression after loading dose of LY2181308. IHC of tumor biopsy from 1 representative patient with breast cancer (A–F) is shown prior to (A, C, E) and following (B, D, F) LY2181308 treatment stained with H&E (1× in A and B; 20× in C and D) and survivin antibody (20× in E, F). G, percent change of survivin mRNA and protein (nuclear and cytoplasmic) expression by IHC with medians and 95% confidence limits. Flow cytometric analysis of survivin expression by endobronchial NSCLC obtained from 3 patients by fiberoptic-guided bronchial brushing prior to and after LY2181308 (H) and change in high survivin-expressing cells (I). PE, phycoerythrin.

Fig. 1.

Reduction of survivin protein and mRNA expression after loading dose of LY2181308. IHC of tumor biopsy from 1 representative patient with breast cancer (A–F) is shown prior to (A, C, E) and following (B, D, F) LY2181308 treatment stained with H&E (1× in A and B; 20× in C and D) and survivin antibody (20× in E, F). G, percent change of survivin mRNA and protein (nuclear and cytoplasmic) expression by IHC with medians and 95% confidence limits. Flow cytometric analysis of survivin expression by endobronchial NSCLC obtained from 3 patients by fiberoptic-guided bronchial brushing prior to and after LY2181308 (H) and change in high survivin-expressing cells (I). PE, phycoerythrin.

Close modal

Survivin protein expression as measured by IHC was reduced by 21%, concomitant with a statistically significant 20% reduction in mRNA expression (P < 0.05) as shown by gene expression analysis (Fig. 1G). Because of the high intensity of nuclear survivin IHC staining prior to the treatment, reduction of nuclear protein expression was more clearly detected than changes seen in the cytoplasm (Fig. 1E–G). Similarly, in the site-specific study in NSCLC, a reduction of survivin protein expression was demonstrated by flow cytometry in endobronchial tumor cells compared with pretreatment levels in all 3 patients studied (Fig. 1I).

The PK of LY2181308 confirmed a multiphasic disposition in plasma with rapid tissue distribution and half-lives of 0.5, 2.5, and 12 hours, and an elimination half-life of 31 days (Fig. 2A). The terminal half-life was the result of a clearance of 20 to 30 L/h and volume of distribution of more than 1,000 L consistent with the tissue distribution of LY2181308 of approximately 90%. As described for other ASOs (9), the primary sites of normal tissue uptake of LY2181308 were renal and hepatic, with less uptake seen in other normal tissue (17). The ASO was detected by IHC in tumor tissue in 10 of 11 patients, either within tumor cells or tumor-associated macrophages (Fig. 2B and C) and in 5 of 11 patients in stroma (Fig. 2D and E). Compatible with the IHC staining, the [11C]LY2181308 PET imaging confirmed that following administration of 1 mg of [11C]LY2181308, tumor penetration of LY2181308 ranged from 21 to 84 ng h/mL tumor with a maximum concentration that ranged from 10 to 60 ng/mL. These levels are comparable with the concentration measured by ELISA in tumor biopsy and with that required for target inhibition (Table 3). The PK profile and tissue distribution of LY2181308 were consistent with the PD effects of the agent described above. However, there was no evident relationship between the localization of the ASO within the tumor tissue compartments, clinical response, or change in survivin expression, perhaps due to the limited numbers of patients included within each cohort.

Fig. 2.

Plasma and tissue PKs following 750-mg dose of LY2181308. Plasma PK following loading and maintenance dosing (A). Observed values (open circles) are depicted with predicted exposures (median 5th and 95th percentiles). Tissue levels of LY2181308 detected by IHC in 2 representative patients (B–E) before (B, D) and following loading dose (C, E) in tumor, 10 of the 11 patients showing a similar pattern (B, C), and in stromal cells, 5 of the 11 patients showing a similar pattern (D, E). Tissue PKs assessed by [11C]LY2181308 uptake (AUC during 90 minutes scan) scaled between the 0 and 140 ng h/mL window for a 1-mg dose: prior to first loading dose (F); and during the second half of the maintenance dose on day 15 (G). Inserts highlight change in uptake in specific area of mesothelioma over time.

Fig. 2.

Plasma and tissue PKs following 750-mg dose of LY2181308. Plasma PK following loading and maintenance dosing (A). Observed values (open circles) are depicted with predicted exposures (median 5th and 95th percentiles). Tissue levels of LY2181308 detected by IHC in 2 representative patients (B–E) before (B, D) and following loading dose (C, E) in tumor, 10 of the 11 patients showing a similar pattern (B, C), and in stromal cells, 5 of the 11 patients showing a similar pattern (D, E). Tissue PKs assessed by [11C]LY2181308 uptake (AUC during 90 minutes scan) scaled between the 0 and 140 ng h/mL window for a 1-mg dose: prior to first loading dose (F); and during the second half of the maintenance dose on day 15 (G). Inserts highlight change in uptake in specific area of mesothelioma over time.

Close modal
Table 3.

Plasma and tissue PKs of LY2181308 at 750 mg

 Predicted 1,000 simulationsObserved n = 24
Plasma PK 
 AUC0–24, ng h/mL 283,725 342,794  
 (133,922–582,041) (187,344–603,944)  
Cmax, ng/mL 65,489 69,120  
 (42,838–96,897) (39923–155,514)  
Cmin, ng/mL 93.8 73.8  
 (63.9–131) (36.8–135.9)  
 1,000 simulations 11C-PET ELISA 
  (n = 4) (n = 5) 
Tumor tissue PKs 
Concentration, ng/mL 33.2 32.5 22.4 
 (18.8–54.0) (13.9–52.8) (3.64–87.4) 
 Predicted 1,000 simulationsObserved n = 24
Plasma PK 
 AUC0–24, ng h/mL 283,725 342,794  
 (133,922–582,041) (187,344–603,944)  
Cmax, ng/mL 65,489 69,120  
 (42,838–96,897) (39923–155,514)  
Cmin, ng/mL 93.8 73.8  
 (63.9–131) (36.8–135.9)  
 1,000 simulations 11C-PET ELISA 
  (n = 4) (n = 5) 
Tumor tissue PKs 
Concentration, ng/mL 33.2 32.5 22.4 
 (18.8–54.0) (13.9–52.8) (3.64–87.4) 

NOTE: Values given are mean (range).

Of 22 patients assessed for efficacy using the RECIST (response evaluation criteria in solid tumors) criteria (20), a total of 4 patients achieved stable disease, including 1 patient with metastatic melanoma who remained free of disease progression for 18 months. Reduction of survivin expression was associated with an increase in the apoptosis marker CC3 and a reduction in tumor expression of the proliferation marker Ki67 (Fig. 3A and B). In the single patient assessed, a partial metabolic response in several mesothelioma lesions was observed with a 40% reduction in standard uptake values (SUV) on FDG-PET imaging (21; Fig. 3C and D).

Fig. 3.

Apoptosis pathway restoration and PD responses in patients receiving 750 mg LY2181308. IHC detection of CC3 (A) and Ki67 (B) in tumor tissue obtained pre- and posttreatment with LY2181308. Percentage change from baseline is represented with medians and interquartile ranges. [18F]FDG-PET images (40–60 minutes) uptake scaled between 0 and 17 g/mL (SUV): prior to first loading dose (C); and following the maintenance dose on day 22 (D). Inserts highlight change in uptake in mesothelioma tumor (same subject as Fig. 2F and G).

Fig. 3.

Apoptosis pathway restoration and PD responses in patients receiving 750 mg LY2181308. IHC detection of CC3 (A) and Ki67 (B) in tumor tissue obtained pre- and posttreatment with LY2181308. Percentage change from baseline is represented with medians and interquartile ranges. [18F]FDG-PET images (40–60 minutes) uptake scaled between 0 and 17 g/mL (SUV): prior to first loading dose (C); and following the maintenance dose on day 22 (D). Inserts highlight change in uptake in mesothelioma tumor (same subject as Fig. 2F and G).

Close modal

Few FHD studies of ASOs have sought to demonstrate PD changes in cancer patients at safe doses such as reduction of the targeted mRNA and protein levels in tumor tissue (22–25). This FHD study of the second-generation ASO LY2181308 was designed to include analysis of serial tumor samples to prove the concept that the ASO was able to inhibit the function of survivin at a safe dose and schedule. We show that the ASO accumulated in tumor tissue, significantly downregulated tumor survivin mRNA and protein expression and enhanced the expression of markers indicative of restored tumor cell apoptosis.

Consistent with previous studies, serial tumor biopsy procedures were safe and accepted by patients (26). Analysis of pre- and posttreatment biopsies demonstrated a reduction of tumor mRNA and protein expression by approximately 20% in a wide range of tumor types. This reduction is lower than that seen in xenograft models, where up to 50% reduction in survivin mRNA and protein were reported (10). In 5 cases, the presence of melanin, bilirubin, or hemosiderin in tumor tissue interfered with the IHC staining and consequently changes in survivin levels after treatment with LY2181308 could not be determined. Tumor samples obtained from patients with NSCLC using fiberoptic bronchoscopy were perhaps even more effective in demonstrating survivin reduction. However, a larger number of patients would need to be included in future studies to confirm our observation. Although survivin levels were reduced, objective clinical responses were not observed and thus direct cytotoxic effects on tumors were difficult to determine. This was an expected finding in the context of a FHD study and because preclinical data suggested that LY2181308 has a cytostatic, rather than a cytotoxic, antitumor effect. Supporting this mode of action were the observations that 1 patient with metastatic melanoma had stable disease while receiving maintenance LY2181308 weekly for 18 months, and 1 patient who had a partial metabolic tumor response based on FDG-PET imaging. Although antitumor efficacy was not a primary endpoint of the study, it was disappointing that no objective responses were seen. Whether this would have been achieved had survivin mRNA and/or protein levels been reduced by more than 20% cannot be addressed by the current study. Combination studies of LY2181308 will assess whether inhibition of survivin enhances antitumor effects of proapoptotic agents such as docetaxel.

An important goal of this study was to demonstrate that PD responses were consistent with the predicted PK profile of LY2181308 in plasma and tissue (Table 3) and its similarity with other second-generation ASOs (9). This prediction was confirmed at the 750 mg dose (Table 3). This is consistent with the observation that ASOs can be scaled successfully from animal to human. The PK profile of LY2181308 and its terminal half-life of about 30 days require a weekly maintenance dose to keep tissue concentrations at levels that were associated with target inhibition in animals. We also demonstrate that the ASO penetrates tumor tissue as detected by IHC and quantitative assessment of ASO levels by ELISA (Table 3). The simulated concentrations in the tumor tissue were confirmed by concentration measurements using ELISA and the [11C]LY2181308 study (Table 3). The range of LY2181308 concentration level observed in the tumor is slightly lower though overlapping relative to the range of LY2181308 through plasma concentration. This could be explained by the fact that other tissues, such as liver and kidney, in addition to the tumor, contribute to the equilibrium between LY2181308 tissues and plasma concentrations The advantage of employing IHC is that it was possible to localize both the disposition of the ASO within tumor tissue and its intracellular distribution. There appeared to be a difference in localization of the ASO within tumor cells and the tumor microenvironment. Whether this observation was a result of the wide range of tumor types studied, the site of the biopsy or the timing after the last dose of LY2181308 is unclear (15 patients had their biopsy taken on day 4; 8 on day 5; 1 on day 6; and 2 on day 7). The variance in timing of posttreatment biopsies resulted in a degree of heterogeneity of results and may have introduced bias. Future studies should define consistent timing of biopsies for all patients and study sites. The use of [11C]LY2181308 PET further supported the IHC-based observation that the ASO penetrated tumor tissue at a pharmacologically relevant concentration. Hence, LY2181308 clearly accumulated in tumor tissue over time (17). This is consistent with studies in animals, where the distribution and stability of the second-generation ASOs was evaluated (9, 27). Other FHD studies evaluating inhibitors of the IAP family have not reported similar PD changes in solid tumor tissue after dosing with ASOs. For example, the small molecular weight survivin inhibitor YM155 was only evaluated for safety and PKs (28), whereas in the clinical trial of the X-linked IAP (XIAP) ASO inhibitor AEG35156 PD changes in PBMCs (peripheral blood mononuclear cells) were reported in addition to safety and PKs (25). Furthermore, the first-generation ASO oblimersen was evaluated for PD activity in melanoma patients only, but the proposed dose and dose schedule was not chosen for future phase 2 studies (22). The second-generation ASO OGX-011 against clusterin did evaluate PD changes in prostate cancer tumor tissue and is the only trial that established a dose and dose regimen based on PD activity in cancer patients that was later used in phase 2 studies (23).

Finally, the dose range at which we observed consistent survivin reduction in tumor tissue had a favorable toxicity profile for future clinical development. Grade 3 and 4 toxicities were present in 11 patients (27.5%) and acute renal failure was not observed during the first cycles of treatment as reported for YM-155, a small molecule survivin inhibitor (28). However, we did observe increased creatinine values (grade 2), which returned to baseline levels after stopping the agent in 1 patient with metastatic melanoma who received LY2157299 at the 750 mg dose for 18 months (29). Although several confounding factors were present, we cannot exclude that LY2157299 treatment was associated with this reversible renal injury. Although not considered medically adverse, lymphopenia was seen in 70% of the patients. Whether this was a PD effect of LY2181308-induced apoptosis of survivin-expressing lymphocytes (30) or reflected an off-target effect of the ASO will require additional investigation. A similar effect on lymphocyte counts was seen with the ASO AEG35156 against XIAP (25). Typical off-target effects of ASO administration were observed for LY2181308 including anemia, thrombocytopenia, and transient prolongation in aPTT (31). In contrast to the phosphothioate ASOs or high doses of second-generation ASOs, the off-target toxicity of LY2181308 was milder and generally limited to grade 1 and 2. This favorable toxicity profile was also recently confirmed in Japanese patients, in particular the loading dose-associated elevation of complement Bb (32).

In conclusion, the integration of PD and PK analyses in this FHD study has provided the proof of concept of effective and specific downregulation of the key molecular target, survivin, in tumor tissue by the second-generation ASO, LY2181308. These findings validate the application of second-generation ASOs for cancer patients. The 750-mg dose and schedule of LY2181308 is currently being evaluated in clinical studies in combination with agents that induce apoptosis such as chemotherapy or radiation (33, 34).

The authors M. Ranson, S. Callies, V. Andr!e, S. Kadam, M. Burgess, and C. Slapak are employees of Eli Lilly and Company, are fully compensated, and hold stock in the company. D. C. Talbot has received other commercial research support, honoraria from Speaker's Bureau, and is on the Advisory Board for Eli Lilly and Company. The other authors declare no potential conflicts of interest.

We thank Drs. C. Prenant, G. Brown, T. Jones, and A. McMahon for [11C]LY2183108 radiolabeling, Dr. M. Slade (Oxford Radcliffe Hospitals) for endobronchial tumor sampling, B. Monia (Isis) for review of the manuscript, and J. Grimes, J. Birkett, C. Leppert, C. Stoner, Helen Desmier and Stacey Maxwell for data validation and study report writing.

The study received funding from Cancer Research UK and the Experimental Cancer Medicine Centres of Oxford, Manchester and London. The study was sponsored by Eli Lilly and Company.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Ambrosini
G
,
Adida
C
,
Altieri
DC
. 
A novel anti-apoptosis gene, survivin, expressed in cancer and lymphoma
.
Nat Med
1997
;
3
:
917
21
.
2.
Altieri
DC
. 
Survivin, versatile modulation of cell division and apoptosis in cancer
.
Oncogene
2003
;
22
:
8581
9
.
3.
Altieri
DC
. 
Validating survivin as a cancer therapeutic target.
Nat Rev Cancer
2003
;
3
:
46
54
.
4.
Li
F
,
Brattain
MG
. 
Role of the survivin gene in pathophysiology
.
Am J Pathol
2006
;
169
:
1
11
.
5.
Altieri
DC
. 
Targeted therapy by disabling crossroad signaling networks: the survivin paradigm
.
Mol Cancer Ther
2006
;
5
:
478
82
.
6.
Nakahara
T
,
Takeuchi
M
,
Kinoyama
I
, et al
YM155, a novel small-molecule survivin suppressant, induces regression of established human hormone-refractory prostate tumor xenografts
.
Cancer Res
2007
;
67
:
8014
21
.
7.
Bennett
FC
,
Ackermann
EJ
,
Cowsert
LM
,
Bennett
FC
,
Ackermann
EJ
,
Cowsert
LM
,
Bennett
FC
, et al
Antisense modulation of survivin expression
.
US patent 7288530 B2
. 
2007
.
8.
Li
F
,
Ackermann
EJ
,
Bennett
CF
, et al
Pleiotropic cell-division defects and apoptosis induced by interference with survivin function
.
Nat Cell Biol
1999
;
1
:
461
6
.
9.
Henry
SP
,
Geary
RS
,
Yu
R
,
Levin
AA
. 
Drug properties of second-generation antisense oligonucleotides: how do they measure up to their predecessors
?
Curr Opin Investig Drugs
2001
;
2
:
1444
9
.
10.
Callies, S, André V, Patel B, et al. Integrated analysis of preclinical data to support the design of the first in man study of LY2181308, a second generation antisense oligonucleotide. Brit J Clin Pharmacol (in press).
11.
Trotti
A
,
Colevas
AD
,
Setser
A
, et al
CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment
.
Semin Radiat Oncol
2003
;
13
:
176
81
.
12.
Bacus
S
,
Flowers
JL
,
Press
MF
,
Bacus
JW
,
McCarty
KS
 Jr.
The evaluation of estrogen receptor in primary breast carcinoma by computer-assisted image analysis
.
Am J Clin Pathol
1988
;
90
:
233
9
.
13.
Knudsen
BS
,
Allen
AN
,
McLerran
DF
, et al
Evaluation of the branched-chain DNA assay for measurement of RNA in formalin-fixed tissues
.
J Mol Diagn
2008
;
10
:
169
76
.
14.
McGill
R
,
Tukey
JW
,
Larsen
WA
. 
Variations of box plots
.
Am Stat
1978
;
32
:
12
6
.
15.
Busse
WW
,
Wanner
A
,
Adams
K
, et al
Investigative bronchoprovocation and bronchoscopy in airway diseases
.
Am J Respir Crit Care Med
2005
;
172
:
807
16
.
16.
Dence
CS
,
Laforest
R
,
Sun
X
,
Sharp
TL
,
Welch
MJ
,
Mach
RH
. 
Radiochemical synthesis, rodent biodistribution and tumor uptake, and dosimetry calculations of [11C] methylated LY2181308
.
Mol Imaging Biol
2010
;
12
:
608
15
.
17.
Saleem
A
,
Ranson
M
,
Callies
S
, et al
Microdosing imaging pharmacokinetic (PK) study of the antisense oligonucleotide (ASO) to survivin (LY2181308) using positron emission tomography (PET): a novel paradigm in clinical drug development
.
J Clin Oncol
2009
.
Abstract 3578
.
18.
Antoch
G
,
Saoudi
N
,
Kuehl
H
, et al
Accuracy of whole-body dual-modality fluorine-18–2-fluoro-2-deoxy-D-glucose positron emission tomography and computed tomography (FDG-PET/CT) for tumor staging in solid tumors: comparison with CT and PET
.
J Clin Oncol
2004
;
22
:
4357
68
.
19.
Roberts
TG
 Jr.
,
Goulart
BH
,
Squitieri
L
, et al
Trends in the risks and benefits to patients with cancer participating in phase 1 clinical trials
.
JAMA
2004
;
292
:
2130
40
.
20.
Therasse
P
,
Arbuck
SG
,
Eisenhauer
EA
, et al
New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada
.
J Natl Cancer Inst
2000
;
92
:
205
16
.
21.
Young
H
,
Baum
R
,
Cremerius
U
, et al
Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations
. 
European Organization for Research and Treatment of Cancer (EORTC) PET Study Group
.
Eur J Cancer
1999
;
35
:
1773
82
.
22.
Jansen
B
,
Wacheck
V
,
Heere-Ress
E
, et al
Chemosensitisation of malignant melanoma by BCL2 antisense therapy
.
Lancet
2000
;
356
:
1728
33
.
23.
Chi
KN
,
Eisenhauer
E
,
Fazli
L
, et al
A phase I pharmacokinetic and pharmacodynamic study of OGX-011, a 2′-methoxyethyl antisense oligonucleotide to clusterin, in patients with localized prostate cancer
.
J Natl Cancer Inst
2005
;
97
:
1287
96
.
24.
Marshall
JL
,
Eisenberg
SG
,
Johnson
MD
, et al
A phase II trial of ISIS 3521 in patients with metastatic colorectal cancer
.
Clin Colorectal Cancer
2004
;
4
:
268
74
.
25.
Dean
E
,
Jodrell
D
,
Connolly
K
, et al
Phase I trial of AEG35156 administered as a 7-day and 3-day continuous intravenous infusion in patients with advanced refractory cancer
.
J Clin Oncol
2009
;
27
:
1660
6
.
26.
Dowlati
A
,
Haaga
J
,
Remick
SC
, et al
Sequential tumor biopsies in early phase clinical trials of anticancer agents for pharmacodynamic evaluation
.
Clin Cancer Res
2001
;
7
:
2971
6
.
27.
Tavitian
B
,
Terrazzino
S
,
Kuhnast
B
, et al
In vivo imaging of oligonucleotides with positron emission tomography
.
Nat Med
1998
;
4
:
467
71
.
28.
Tolcher
AW
,
Mita
A
,
Lewis
LD
, et al
Phase I and pharmacokinetic study of YM155, a small-molecule inhibitor of survivin
.
J Clin Oncol
2008
;
26
:
5198
203
.
29.
Herrington
WG
,
Talbot
DC
,
Lahn
MM
, et al
Long-term administration of the survivin mRNA-targeted antisense oligonucleotide LY2181308 is associated with reversible kidney injury in a patient with metastatic melanoma
.
Am J Kidney Dis
in press
.
30.
Andersen
MH
,
Svane
IM
,
Becker
JC
,
Straten
PT
. 
The universal character of the tumor-associated antigen survivin
.
Clin Cancer Res
2007
;
13
:
5991
4
.
31.
Jason
TL
,
Koropatnick
J
,
Berg
RW
. 
Toxicology of antisense therapeutics
.
Toxicol Appl Pharmacol
2004
;
201
:
66
83
.
32.
Tanioka
M
,
Nokihara
H
,
Yamamoto
N
, et al
Phase I study of LY2181308, an antisense oligonucleotide against survivin, in patients with advanced solid tumors
.
Cancer Chemother Pharmacol
. 
2010 Nov 16
. [
Epub ahead of print], DOI
: .
33.
Rodel
F
,
Frey
B
,
Leitmann
W
,
Capalbo
G
,
Weiss
C
,
Rodel
C
. 
Survivin antisense oligonucleotides effectively radiosensitize colorectal cancer cells in both tissue culture and murine xenograft models
.
Int J Radiat Oncol Biol Phys
2008
;
71
:
247
55
.
34.
Giodini
A
,
Kallio
MJ
,
Wall
NR
, et al
Regulation of microtubule stability and mitotic progression by survivin
.
Cancer Res
2002
;
62
:
2462
7
.

Supplementary data