Abstract
Lessons Learned.
The addition of the heat shock protein 27 (Hsp27)‐targeting antisense oligonucleotide, apatorsen, to a standard first‐line chemotherapy regimen did not result in improved survival in unselected patients with metastatic pancreatic cancer.
Findings from this trial hint at the possible prognostic and predictive value of serum Hsp27 that may warrant further investigation.
Background.
This randomized, double‐blinded, phase II trial evaluated the efficacy of gemcitabine/nab‐paclitaxel plus either apatorsen, an antisense oligonucleotide targeting heat shock protein 27 (Hsp27) mRNA, or placebo in patients with metastatic pancreatic cancer.
Methods.
Patients were randomized 1:1 to Arm A (gemcitabine/nab‐paclitaxel plus apatorsen) or Arm B (gemcitabine/nab‐paclitaxel plus placebo). Treatment was administered in 28‐day cycles, with restaging every 2 cycles, until progression or intolerable toxicity. Serum Hsp27 levels were analyzed at baseline and on treatment. The primary endpoint was overall survival (OS).
Results.
One hundred thirty‐two patients were enrolled, 66 per arm. Cytopenias and fatigue were the most frequent grade 3/4 treatment‐related adverse events for both arms. Median progression‐free survival (PFS) and OS were 2.7 and 5.3 months, respectively, for arm A, and 3.8 and 6.9 months, respectively, for arm B. Objective response rate was 18% for both arms. Patients with high serum level of Hsp27 represented a poor‐prognosis subgroup who may have derived modest benefit from addition of apatorsen.
Conclusion.
Addition of apatorsen to chemotherapy does not improve outcomes in unselected patients with metastatic pancreatic cancer in the first‐line setting, although a trend toward prolonged PFS and OS in patients with high baseline serum Hsp27 suggests this therapy may warrant further evaluation in this subgroup.
Abstract
经验总结
• 在非经选转移性胰腺癌患者的标准一线化疗方案中添加靶向热休克蛋白27 (Hsp27) 的反义寡核苷酸apatorsen不会延长生存期。
•本试验的结果提示血清Hsp27可能具有预后和预测价值, 但需进行进一步研究。
摘要
背景.本项随机、双盲、II期试验评价了吉西他滨/白蛋白结合型紫杉醇联合apatorsen[靶向热休克蛋白27 (Hsp27) mRNA的反义寡核苷酸]或安慰剂治疗转移性胰腺癌患者的效果。
方法.患者按1:1的比例被随机分配进入A组(吉西他滨/白蛋白结合型紫杉醇联合apatorsen)或B组(吉西他滨/白蛋白结合型紫杉醇联合安慰剂)。治疗周期为28天, 每2个周期对疾病进行一次重新分期, 直至出现疾病进展或不可耐受的毒性。在基线期和治疗期间分析了血清Hsp27水平。主要终点为总生存期(OS)。
结果.共入组了132例患者, 每组66例。血细胞减少和疲乏是两组中最常见的3/4级治疗相关不良事件。A组的中位无进展生存期(PFS)和OS分别为2.7和5.3个月;B组分别为3.8和6.9个月。两组的客观缓解率均为18%。Hsp27血清水平较高的患者代表可能已经从添加apatorsen中适度获益的预后不佳亚组。
结论.在非经选转移性胰腺癌患者的一线化疗中添加apatorsen不会改善治疗结局, 但基线期血清Hsp27水平较高患者的PFS和OS延长的趋势表明可能需在该亚组中对这一疗法进行进一步研究。
Discussion
Heat shock protein 27 (Hsp27) is a protein chaperone whose expression is induced by cytotoxic chemotherapy, as well as other cell stressors such as hyperthermia, oxidative stress, and radiation, resulting in cytoprotection against these insults [1], [2]. Various malignancies, including pancreatic adenocarcinoma, overexpress Hsp27 [3]; furthermore, preclinical studies show that Hsp27 expression may play a role in the resistance of pancreatic cancer cell lines to gemcitabine [4], [5], [6]. Thus, inhibiting Hsp27 activity represents a viable therapeutic strategy in this disease. Apatorsen, an antisense oligonucleotide designed to bind to Hsp27 mRNA and block its translation into functional protein, offers one such approach [7].
On these bases, we performed a randomized phase II trial to compare the safety and efficacy of gemcitabine and nab‐paclitaxel in combination with either apatorsen or placebo in the first‐line setting for patients with metastatic pancreatic cancer. The planned sample size of 130 provided 80% power to detect a difference in median survival of 8.5 versus 13.4 months (hazard ratio [HR] for death, 0.634; 1‐sided a = 0.1).
Table 1. Median PFS and OS stratified by heat shock protein 27 levels.
Abbreviations: CI, confidence interval; Hsp27, heat shock protein 27; OS, overall survival; PFS, progression‐free survival.
Although the incidence of toxicities did not differ significantly between the two treatment arms (most common treatment‐related toxicities of all grades on both arms included fatigue, cytopenias, and gastrointestinal symptoms), the addition of apatorsen to chemotherapy did not produce any improvement in clinical outcomes in the intent‐to‐treat population. The objective response rate (ORR) was identical (18%) on both treatment arms, whereas patients on the apatorsen arm fared numerically worse in terms of both progression‐free survival (PFS) and overall survival (OS) when compared with patients on the placebo arm, although these differences were not statistically significant (median PFS, 2.7 vs. 3.8 months, respectively [p = .92; HR 1.0]; median OS, 5.3 vs. 6.9 months, respectively [p = .62; HR 1.1]). Notably, the survival outcomes for patients on both arms of this study were considerably inferior to those observed on the gemcitabine/nab‐paclitaxel arm from the phase III Metastatic Pancreatic Adenocarcinoma Clinical Trial (MPACT) study [8], for unclear reasons.
The only subgroup for whom a potential benefit of apatorsen could be identified was those patients with high serum levels of Hsp27, a marker associated with a very poor prognosis overall (median PFS for patients with high baseline serum Hsp27 levels, 3.3 vs. 0.9 months for apatorsen vs. placebo, respectively [HR 0.38], median OS 3.3 vs. 1.0 months [HR 0.59]). However, the number of subjects who fit into this category was too small, representing only 14% of the entire study population, to draw any definitive conclusions.
In summary, the addition of apatorsen to a standard combination chemotherapy regimen in the first‐line setting did not result in improvement in survival or other clinically relevant endpoints in patients with metastatic pancreatic cancer. Although further studies of this agent in unselected patients do not appear to be indicated, the findings from this trial do hint at the possible prognostic and predictive value of serum Hsp27 that may warrant further investigation.
Trial Information
- Disease
Metastatic pancreatic adenocarcinoma
- Stage of Disease/Treatment
Metastatic/advanced
- Prior Therapy
None
- Type of Study – 1
Phase II
- Type of Study – 2
Randomized
- PFS
p = 0.92, HR: 1.0
- OS
p = 0.62, HR: 1.1
- Primary Endpoint
Overall survival
- Secondary Endpoint
Progression‐free survival
- Secondary Endpoint
Toxicity
- Investigator's Analysis
Feasible, possibly effective in patients with high Hsp27 serum levels
Drug Information for Phase II Control
- Drug 1
- Generic/Working name
Nab‐paclitaxel
- Trade name
Abraxane
- Company name
Celgene
- Drug type
Other
- Drug class
Microtubule‐targeting agent
- Dose
125 milligrams (mg) per square meter (m2)
- Route
IV
- Schedule of administration
Placebo days 1, 8, 15, and 22 of a 28‐day cycle Nab‐paclitaxel and gemcitabine days 1, 8, and 15 of a 28‐day cycle
- Drug 2
- Generic/Working name
Gemcitabine
- Trade name
Gemzar
- Company name
Eli Lilly
- Drug type
Other
- Drug class
Antimetabolite
- Dose
1,000 mg/m2
- Route
IV
- Schedule of administration
Placebo days 1, 8, 15, and 22 of a 28‐day cycle Nab‐paclitaxel and gemcitabine days 1, 8, and 15 of a 28‐day cycle
Drug Information for Phase II Experimental
- Drug 1
- Generic/Working name
Apatorsen
- Trade name
Not applicable
- Company name
OncoGenex
- Drug type
Other
- Drug class
Antisense oligonucleotide
- Dose
600 mg per flat dose
- Route
IV
- Schedule of administration
Apatorsen days 1, 8, 15, and 22 of a 28‐day cycle Nab‐paclitaxel and gemcitabine days 1, 8, and 15 of a 28‐day cycle
- Drug 2
- Generic/Working name
Nab‐paclitaxel
- Trade name
Abraxane
- Company name
Celgene
- Drug type
Other
- Drug class
Microtubule‐targeting agent
- Dose
125 mg/m2
- Schedule of administration
Apatorsen days 1, 8, 15, and 22 of a 28‐day cycle Nab‐paclitaxel and gemcitabine days 1, 8, and 15 of a 28‐day cycle pfof
- Drug 3
- Generic/Working name
Gemcitabine
- Trade name
Gemzar
- Company name
Eli Lilly
- Drug type
Other
- Drug class
Antimetabolite
- Dose
1,000 mg/m2
- Route
IV
- Schedule of administration
Apatorsen 1, 8, 15, and 22 of a 28‐day cycle Nab‐paclitaxel and gemcitabine days 1, 8, and 15 of a 28‐day cycle
Patient Characteristics for Phase II Control
- Number of Patients, Male
38
- Number of Patients, Female
28
- Stage
IV
- Age
Median (range): 65.5 (47–83)
- Number of Prior Systemic Therapies
Median (range): None
- Performance Status ECOG
-
0 — 32
1 — 32
2 —
3 —
Unknown — 2
- Cancer Types or Histologic Subtypes
-
CA19‐9 levels ≤90 U/mL 11
CA19‐9 levels >90 U/mL 54
CA19‐9 levels Unknown 1
Hsp27 expression high 11
Hsp27 expression low 47
Hsp27 expression Unknown 8
Patient Characteristics for Phase II Experimental
- Number of Patients, Male
37
- Number of Patients, Female
29
- Stage
IV
- Age
Median (range): 66.5 (39–82)
- Number of Prior Systemic Therapies
Median (range): None
- Performance Status: ECOG
-
0 — 30
1 — 36
2 —
3 —
Unknown —
- Cancer Types or Histologic Subtypes
-
CA19‐9 levels ≤90 U/mL 18
CA19‐9 levels >90 U/mL 47
CA19‐9 levels unknown 1
Hsp27 expression high 7
Hsp27 expression low 55
Hsp27 expression unknown 4
Primary Assessment Method for Phase II Control
- Assessment: Total Patient Population: Overall Survival
- Number of patients screened
66
- Number of patients enrolled
66
- Number of patients evaluable for toxicity
63
- Number of patients evaluated for efficacy
66
- Evaluation method
RECIST 1.1
- Response assessment CR
n = 0 (0%)
- Response assessment PR
n = 12 (18%)
- Response assessment SD
n = 21 (32%)
- Response assessment PD
n = 18 (28%)
- Response assessment OTHER
n = 15 (22%)
- (Median) duration assessments PFS
3.8 months
- (Median) duration assessments OS
6.9 months
- Kaplan‐Meier time units
months
Waterfall plot demonstrating best objective response in response‐evaluable subjects on control arm.


Assessment: Total Patient Population: Progression‐Free Survival
- Number of patients screened
66
- Number of patients enrolled
66
- Number of patients evaluable for toxicity
63
- Number of patients evaluated for efficacy
66
- Evaluation method
RECIST 1.1
- Response assessment CR
n = 0 (0%)
- Response assessment PR
n = 12 (18%)
- Response assessment SD
n = 21 (32%)
- Response assessment PD
n = 18 (28%)
- Response assessment OTHER
n = 15 (22%)
- (Median) duration assessments PFS
3.8 months
- (Median) duration assessments OS
6.9 months
- Kaplan‐Meier time units
months

Primary Assessment Method for Phase II Experimental
- Assessment: Total Patient Population: Overall Survival
- Number of patients screened
66
- Number of patients enrolled
66
- Number of patients evaluable for toxicity
64
- Number of patients evaluated for efficacy
66
- Evaluation method
RECIST 1.1
- Response assessment CR
n = 0 (0%)
- Response assessment PR
n = 12 (18%)
- Response assessment SD
n = 16 (24%)
- Response assessment PD
n = 21 (32%)
- Response assessment OTHER
n = 17 (26%)
- (Median) duration assessments PFS
2.7 months
- (Median) duration assessments OS
5.3
- Kaplan-Meier time units
months
Waterfall plot demonstrating best objective response in response‐evaluable subjects on experimental arm.


Assessment: Total Patient Population: Progression‐Free Survival
- Number of patients screened
66
- Number of patients enrolled
66
- Number of patients evaluable for toxicity
64
- Number of patients evaluated for efficacy
66
- Evaluation method
RECIST 1.1
- Response assessment CR
n = 0 (0%)
- Response assessment PR
n = 12 (18%)
- Response assessment SD
n = 16 (24%)
- Response assessment PD
n = 21 (32%)
- Response assessment OTHER
n = 17 (26%)
- Kaplan-Meier time units
months

Adverse Events
Assessment, Analysis, and Discussion
- Completion
Study completed
- Pharmacokinetics/Pharmacodynamics
Correlative endpoints not met
- Investigator's Assessment
Feasible, possibly effective in patients with high Hsp27 serum levels
Pancreatic adenocarcinoma is expected to rise to the second leading cause of cancer‐related mortality in the U.S. by the end of this decade [9]. Systemic therapy represents the mainstay of treatment for patients with advanced or metastatic disease, for whom two combination chemotherapy regimens have emerged as front‐line standards of care: gemcitabine plus nab‐paclitaxel [8], and FOLFIRINOX (5‐fluorouracil, leucovorin, irinotecan, and oxaliplatin) [10]. However, despite these recent improvements in available cytotoxic therapies, overall prognosis in this disease remains very poor; for example, in the phase III MPACT trial leading to the approval of nab‐paclitaxel for metastatic pancreatic cancer, the median survival for patients receiving the combination of gemcitabine plus nab‐paclitaxel was only 8.5 months, representing a statistically significant but relatively modest absolute improvement of 1.8 months when compared with single‐agent gemcitabine [8]. Clearly, novel drugs with unique mechanisms of action warrant further exploration.
Molecularly targeted therapies that have been approved for use in clinical oncology include monoclonal antibodies and small molecule inhibitors, particularly tyrosine kinase inhibitors. Unfortunately, many potential therapeutic targets are not amenable to these specific pharmacologic approaches, highlighting the importance of developing alternative strategies, including agents that can disrupt these targets at the gene expression level. Antisense oligonucleotides (ASOs), which are chemically modified stretches of single‐strand DNA complementary to the mRNA regions of a target gene that inhibit translation by forming RNA/DNA duplexes, represent one such approach to specifically prevent translation of functionally relevant genes.
Apatorsen, a 2’‐methoxyethyl oligonucleotide with phosphorothiolated internucleotide linkages, is designed to bind to heat shock protein 27 (Hsp27) mRNA and prevent translation into a functional protein. Targeting this specific heat shock protein represents an attractive therapeutic option, because Hsp27 can potentially affect multiple pathways implicated in cancer progression and resistance, as opposed to targeting a single pathway, a strategy that might have limited benefits in the face of the redundant signaling pathways and significant tumor heterogeneity. For example, overexpression of Hsp27 in cancer cells is induced by cytotoxic chemotherapy, as well as other cell stressors including hyperthermia, oxidative stress, and radiation, resulting in cytoprotection against these insults [1], [2]. Furthermore, Hsp27 serves to stabilize mutated or inappropriately activated oncoproteins that contribute to the initiation, growth, and metastasis of human cancers [2], [11].
The phase I dose‐escalation study of apatorsen in patients with castration‐resistant prostate cancer and other advanced cancers showed evidence of monotherapy activity as demonstrated by decline in tumor markers and circulating tumor cells, as well as stable measurable disease in 12 of 42 patients [12]. Further evaluation of apatorsen has been pursued in combination with chemotherapeutic agents in clinical trials specific to non‐small cell lung and bladder cancer [13], [14].
Pancreatic adenocarcinomas show higher levels of Hsp27 expression when compared with healthy pancreatic tissue, and the protein can also be detected with high sensitivity in the serum of patients with pancreatic cancer [3]. Preclinical studies additionally show that Hsp27 expression may play a role in the resistance of pancreatic cancer cell lines to gemcitabine [4], [5], [6]. On these bases, the current study was designed to compare the safety and efficacy of gemcitabine and nab‐paclitaxel in combination with either apatorsen or placebo in the first‐line setting for patients with metastatic pancreatic cancer. However, we did not observe any improvement in clinical outcomes in the intent‐to‐treat population in this trial; indeed, patients on the apatorsen arm fared numerically worse in terms of both PFS and OS when compared with patients on the placebo control arm, although these differences were not statistically significant. The only subgroup for whom a potential benefit of apatorsen could be identified was those patients with high serum levels of Hsp27, a marker that portended a very poor prognosis overall. However, the number of subjects who fit into this category was too small, representing only 14% of the entire study population, to draw any definitive conclusions. Further studies, either prospectively designed trials or retrospective analyses of available clinically annotated samples, will be required to assess whether Hsp27 truly represents an adverse prognostic marker in this disease.
Several additional points are worth noting. First, the clinical outcomes for patients on both arms of this study were considerably inferior to those observed on the gemcitabine plus nab‐paclitaxel arm from the phase III MPACT trial [8], in which patients achieved a median OS and PFS of 8.5 and 5.5 months, respectively. It is unclear why subjects on the current study fared so poorly, given similar demographics to the MPACT study; nevertheless, it seems fairly unlikely that a benefit from apatorsen would somehow be unmasked even if patients had achieved outcomes that more similarly matched those of the MPACT trial. Furthermore, because this study did not mandate pretreatment collection of tumor specimens, putative tissue‐based predictive markers of apatorsen sensitivity could not be assessed, nor could the pharmacodynamic effects of this agent given the absence of requiring on‐treatment tumor biopsies. No consistent trend was identified on serial serum Hsp27 measurements in responders versus nonresponders (data not shown). This lack of robust correlative data represents a major limitation of our study and highlights one of the major ongoing challenges in pancreatic cancer trial design in general, especially when trying to confirm the putative mechanism of action of novel targeted agents.
In conclusion, the addition of apatorsen to a standard combination chemotherapy regimen in the first‐line setting did not result in improved survival or other clinically relevant endpoints in patients with metastatic pancreatic cancer. Further studies of this agent in unselected patients do not appear to be indicated, although the findings from this trial do hint at the possible prognostic and predictive value of serum Hsp27 that may warrant further investigation. If ASO technologies targeting this and other cancer‐related genes continue to be pursued in pancreatic cancer, they should ideally be evaluated in the context of trials that mandate serial collection of both tumor and blood samples to look for predictive markers and pharmacodynamic markers of response, notwithstanding the clinical and logistic hurdles these may present in this patient population.
Acknowledgments
This work was supported in part by a grant from OncoGenex Pharmaceuticals, Inc.
Footnotes
ClinicalTrials.gov Identifier: NCT01844817
Sponsor: Sarah Cannon Research Institute
Principal Investigators: Andrew H. Ko, Johanna C. Bendell
IRB Approved: Yes
See the related commentary on page 1424.
Disclosures
Andrew H. Ko: Seattle Genetics, New Beta Innovations (C/A), Merck, Bristol‐Myers Squibb, Abgenomics, Prism Bio Ltd, Merrimack, Halozyme, Celgene, Roche/Genentech (RF); David M. Waterhouse: Bristol‐Myers Squibb, ABBVie (H). The other authors indicated no financial relationships.
(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board
References
- 1. Garrido C, Brunet M, Didelot C et al. Heat shock proteins 27 and 70: Anti‐apoptotic proteins with tumorigenic properties. Cell Cycle 2006;5:2592–2601. [DOI] [PubMed] [Google Scholar]
- 2. Ciocca DR, Calderwood SK. Heat shock proteins in cancer: Diagnostic, prognostic, predictive, and treatment implications. Cell Stress Chaperones 2005;10:86–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Melle C, Ernst G, Escher N et al. Protein profiling of microdissected pancreas carcinoma and identification of HSP27 as a potential serum marker. Clin Chem 2007;53:629–635. [DOI] [PubMed] [Google Scholar]
- 4. Kuramitsu Y, Wang Y, Taba K et al. Heat‐shock protein 27 plays the key role in gemcitabine‐resistance of pancreatic cancer cells. Anticancer Res 2012;32:2295–2299. [PubMed] [Google Scholar]
- 5. Zhang S, Zhang XQ, Huang SL et al. The effects of HSP27 on gemcitabine‐resistant pancreatic cancer cell line through snail. Pancreas 2015;44:1121–1129. [DOI] [PubMed] [Google Scholar]
- 6. Guo Y, Ziesch A, Hocke S et al. Overexpression of heat shock protein 27 (HSP27) increases gemcitabine sensitivity in pancreatic cancer cells through S‐phase arrest and apoptosis. J Cell Mol Med 2015;19:340–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Kamada M, So A, Muramaki M et al. Hsp27 knockdown using nucleotide‐based therapies inhibit tumor growth and enhance chemotherapy in human bladder cancer cells. Mol Cancer Ther 2007;6:299–308. [DOI] [PubMed] [Google Scholar]
- 8. Von Hoff DD, Ervin T, Arena FP et al. Increased survival in pancreatic cancer with nab‐paclitaxel plus gemcitabine. N Engl J Med 2013;369:1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Rahib L, Smith BD, Aizenberg R et al. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 2014;74:2913–2921. [DOI] [PubMed] [Google Scholar]
- 10. Conroy T, Desseigne F, Ychou M et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817–1825. [DOI] [PubMed] [Google Scholar]
- 11. Acunzo J, Andrieu C, Baylot V et al. Hsp27 as a therapeutic target in cancers. Curr Drug Targets 2014;15:423–431. [DOI] [PubMed] [Google Scholar]
- 12. Chi KN, Yu EY, Jacobs C et al. A phase I dose‐escalation study of apatorsen (OGX‐427), an antisense inhibitor targeting heat shock protein 27 (Hsp27), in patients with castration‐resistant prostate cancer and other advanced cancers. Ann Oncol 2016;27:1116–1122. [DOI] [PubMed] [Google Scholar]
- 13. Choueiri TK, Hahn NM, Pal SK et al. The Borealis‐2 clinical trial: A randomized phase 2 study of OGX‐427 (apatorsen) plus docetaxel versus docetaxel alone in relapsed/refractory metastatic urothelial cancer J Clin Oncol 2014;32; (suppl):TPS4593a. [Google Scholar]
- 14. Spigel DR, Burris HA, Greco FA et al. Double‐blind randomized phase II trial of carboplatin and pemetrexed with or without OGX‐427 in patients with previously untreated stage IV non‐squamous non‐small‐cell lung cancer (NSCLC): The Spruce Clinical Trial J Clin Oncol 2013;31(suppl):TPS8120a. [DOI] [PMC free article] [PubMed] [Google Scholar]






