Oncolytic adenovirus with temozolomide induces autophagy and antitumor immune responses in cancer patients - PubMed (original) (raw)

. 2013 Jun;21(6):1212-23.

doi: 10.1038/mt.2013.51. Epub 2013 Apr 2.

Laura Ahtiainen, Mari L M Hirvinen, Simona Bramante, Vincenzo Cerullo, Petri Nokisalmi, Otto Hemminki, Iulia Diaconu, Sari Pesonen, Anniina Koski, Lotta Kangasniemi, Saila K Pesonen, Minna Oksanen, Leena Laasonen, Kaarina Partanen, Timo Joensuu, Fang Zhao, Anna Kanerva, Akseli Hemminki

Affiliations

Oncolytic adenovirus with temozolomide induces autophagy and antitumor immune responses in cancer patients

Ilkka Liikanen et al. Mol Ther. 2013 Jun.

Abstract

Oncolytic adenoviruses and certain chemotherapeutics can induce autophagy and immunogenic cancer cell death. We hypothesized that the combination of oncolytic adenovirus with low-dose temozolomide (TMZ) is safe, effective, and capable of inducing antitumor immune responses. Metronomic low-dose cyclophosphamide (CP) was added to selectively reduce regulatory T-cells. Preclinically, combination therapy inhibited tumor growth, increased autophagy, and triggered immunogenic cell death as indicated by elevated calreticulin, adenosine triphosphate (ATP) release, and nuclear protein high-mobility group box-1 (HMGB1) secretion. A total of 41 combination treatments given to 17 chemotherapy-refractory cancer patients were well tolerated. We observed anti- and proinflammatory cytokine release, evidence of virus replication, and induction of neutralizing antibodies. Tumor cells showed increased autophagy post-treatment. Release of HMGB1 into serum--a possible indicator of immune response--increased in 60% of treatments, and seemed to correlate with tumor-specific T-cell responses, observed in 10/15 cases overall (P = 0.0833). Evidence of antitumor efficacy was seen in 67% of evaluable treatments with a trend for increased survival over matched controls treated with virus only. In summary, the combination of oncolytic adenovirus with low-dose TMZ and metronomic CP increased tumor cell autophagy, elicited antitumor immune responses, and showed promising safety and efficacy.

PubMed Disclaimer

Figures

Figure 1

Figure 1

Immunogenic cell killing and increased autophagy coincide with tumor growth inhibition in oncolytic adenovirus, temozolomide (TMZ)- and cyclophosphamide-treated prostate cancer. (a) Cell-killing efficacy of combination treatments. Ten virus particles (VP)/cell of oncolytic adenovirus combined with TMZ (c1 = 0.035, c2 = 0.105 mg/ml) and 4-HPCP (c1 = 0.003125, c2 = 0.00525 mg/ml) resulted in superior cell killing over chemotherapeutic agents or virus alone (P < 0.01,P < 0.05, respectively). (b) Immunogenicity of cell death. Combination treatment with 100 VP/cell of Ad5/3-D24-GMCSF virus, TMZ (0.0025 mg/ml), and 4-HPCP (0.00208 mg/ml) resulted in significant increase in calreticulin-positive PC3-MM2 cells, and extracellular ATP and HMGB1 levels, as compared with untreated cells. Treatment with TMZ and 4-HPCP increased calreticulin-positive cells and ATP release, whereas oncolytic virus seemed to induce mostly ATP and HMGB1 release, but lacked significant induction of others. Data ina,b are representative of three independent experiments. (c) Efficacy of combination therapy in vivo. Nude/NMRI mice bearing subcutaneous PC3-MM2 xenografts were treated intratumorally with Ad5/3-D24-GMCSF virus or growth medium (black arrows) followed by intraperitoneal injections of TMZ or saline (gray arrows), and cyclophosphamide (CP) or saline (white arrowheads). Virus + TMZ, and virus + TMZ + CP treatments significantly inhibited tumor growth as compared with phosphate-buffered saline (PBS) control. (d,e) Induction of autophagy after combination therapy in vivo. (d) Electron microscopy on fixed tumor tissues revealed large tumor cells with enlarged nuclei, abundant mitochondria, ribosomes, and glycogen deposits (gray vacuoles). Autophagosomes and autolysosomes were only found in virus-, virus + TMZ-, and virus + TMZ + CP-treated tumor cells (black arrowheads). VPs were observed inside some of the autophagic cells (white arrowheads). (e) Tumors were assessed by immunohistochemistry for LC3, a membrane-bound protein accumulating on late autophagosomes. Punctate staining pattern was considered indicative of autophagy (black arrowheads; ×40 original magnification). Error bars represent the mean ± SEM. All studies, n = 3–6; *P < 0.05; **P < 0.01; ***P < 0.001; unpaired _t_-tests ina,b; one-way analysis of variance repeated measures in c. 4-HPCP, 4-hydroperoxycyclophosphamide; ATP, adenosine triphosphate; GMCSF, granulocyte-macrophage colony-stimulating factor; HMGB1, high-mobility group box-1.

Figure 2

Figure 2

Computed tomography of an endometrial sarcoma patient, and autophagy induction in patient ascites tumor cells after combination treatment. (ac) Metastases near the pancreas (white arrowheads) (a) before treatment, and injected target lesions (b) after the first round of oncolytic virotherapy together with metronomic cyclophosphamide (CP), and temozolomide (TMZ) for 5 days before virus resulted in 5% decrease in tumor diameters. (c) Second treatment with Ad5/3-D24-GMCSF, metronomic CP, and TMZ for 5 days before virus resulted in 15% reduction. (d) Pre- and post-treatment ascites cells of two pancreatic carcinoma patients treated with Ad5/3-hTERT-E1A-CD40L virus, metronomic CP, and TMZ for 7 days after virus were assessed for autophagy by LC3 immunohistochemistry. Higher frequency of LC3 punctate-positive tumor cells (black arrowheads) was observed in post-treatment samples (right column) indicative of autophagy induction. No clinical information was available for these patients and therefore they are not included in the other display items. GMCSF, granulocyte-macrophage colony-stimulating factor.

Figure 3

Figure 3

Antitumor T-cells are stimulated by combination treatments and correlate with HMGB1 secretion into serum; a potential marker of immunogenic cell death. (a) Peripheral blood mononuclear cells (PBMCs) were pulsed with peptide mix for adenovirus serotype 5 (Ad5) penton (gray bars) or for survivin (black bars), a ubiquitous tumor-associated antigen, and assessed for T-cell activation by interferon-γ ELISPOT analysis. Unspecific T-cell responses were also observed, which might include other tumor epitope-directed T-cells (white bars). Induction of antitumor T-cells was observed in eight cases (open triangles) and decrease/trafficking in two cases (closed triangles). Adenovirus-specific T-cell activations were seen in 10/15 cases (open circles). Patient S119 received temozolomide (TMZ) according to group 1 and later group 3; for convenience, both are shown in the group 3 row. The adenoviral peptide mix was for serotype 3 in the latter case (Ad3 hexon). (b) Quantitative determination of HMGB1 in patient serum was performed by enzyme-linked immunosorbent assay. Significant elevations of the immunogenic signal over baseline were observed in 60% of patients (*P < 0.05; **P < 0.01; unpaired _t_-test). HMGB1 titer change correlated with antitumor T-cell responses (triangle markers) in 8/9 evaluable cases. The only exception was patient C145, who showed a high antitumor T-cell induction at week 4 post-treatment, but lacked HMGB1 elevation at the time (triangle in parenthesis). The nine evaluable cases were S149, N127, M3, M50, C145, K152, and S171 2nd, S119 1st, and S119 2nd treatment rounds; Patient C156 had absolute T-cell counts below the limit of reliable detection and was therefore excluded from statistical analyses. Error bars represent the mean ± SEM, n = 3 per timepoint. 1st, initial treatment rounds; 2nd, later treatment rounds; Group 1, TMZ before virus; Group 2, TMZ before and after virus; Group 3, TMZ after virus; HMGB1, high-mobility group box-1; SFC, spot-forming colonies.

Figure 4

Figure 4

Survival of combination-treated patients is increased over matched nonrandomized control patients. Overall survival of 17 oncolytic adenovirus, temozolomide (TMZ)- and cyclophosphamide (CP)-treated patients was analyzed by the Kaplan–Meier method. Matching control patients (n = 17) treated without TMZ were selected retrospectively from the same patient cohort taking into account putative prognostic factors. Median survival in the combination-treated (Virus + TMZ) patients was 269 days versus 170 days in virus-treated controls. One combination-treated patient was alive at the end of follow-up with a survival of 1,459 days.

Similar articles

Cited by

References

    1. Ito H, Aoki H, Kühnel F, Kondo Y, Kubicka S, Wirth T, et al. Autophagic cell death of malignant glioma cells induced by a conditionally replicating adenovirus. J Natl Cancer Inst. 2006;98:625–636. - PubMed
    1. Jiang H, White EJ, Ríos-Vicil CI, Xu J, Gomez-Manzano C, Fueyo J. Human adenovirus type 5 induces cell lysis through autophagy and autophagy-triggered caspase activity. J Virol. 2011;85:4720–4729. - PMC - PubMed
    1. Chen N, Karantza V. Autophagy as a therapeutic target in cancer. Cancer Biol Ther. 2011;11:157–168. - PMC - PubMed
    1. Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 2011;334:1573–1577. - PubMed
    1. Martins I, Michaud M, Sukkurwala AQ, Adjemian S, Ma Y, Shen S, et al. Premortem autophagy determines the immunogenicity of chemotherapy-induced cancer cell death. Autophagy. 2012;8:413–415. - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources