Skip to main content
Top
Published in: BMC Cancer 1/2017

Open Access 01-12-2017 | Research article

Photodynamic therapy combined to cisplatin potentiates cell death responses of cervical cancer cells

Authors: Laura Marise de Freitas, Rodolfo Bortolozo Serafim, Juliana Ferreira de Sousa, Thaís Fernanda Moreira, Cláudia Tavares dos Santos, Amanda Martins Baviera, Valeria Valente, Christiane Pienna Soares, Carla Raquel Fontana

Published in: BMC Cancer | Issue 1/2017

Login to get access

Abstract

Background

Photodynamic therapy (PDT) has proven to be a promising alternative to current cancer treatments, especially if combined with conventional approaches. The technique is based on the administration of a non-toxic photosensitizing agent to the patient with subsequent localized exposure to a light source of a specific wavelength, resulting in a cytotoxic response to oxidative damage. The present study intended to evaluate in vitro the type of induced death and the genotoxic and mutagenic effects of PDT alone and associated with cisplatin.

Methods

We used the cell lines SiHa (ATCC® HTB35™), C-33 A (ATCC® HTB31™) and HaCaT cells, all available at Dr. Christiane Soares’ Lab. Photosensitizers were Photogem (PGPDT) and methylene blue (MBPDT), alone or combined with cisplatin. Cell death was accessed through Hoechst and Propidium iodide staining and caspase-3 activity. Genotoxicity and mutagenicity were accessed via flow cytometry with anti-gama-H2AX and micronuclei assay, respectively. Data were analyzed by one-way ANOVA with Tukey’s posthoc test.

Results

Both MBPDT and PGPDT induced caspase-independent death, but MBPDT induced the morphology of typical necrosis, while PGPDT induced morphological alterations most similar to apoptosis. Cisplatin predominantly induced apoptosis, and the combined therapy induced variable rates of apoptosis- or necrosis-like phenotypes according to the cell line, but the percentage of dead cells was always higher than with monotherapies. MBPDT, either as monotherapy or in combination with cisplatin, was the unique therapy to induce significant damage to DNA (double strand breaks) in the three cell lines evaluated. However, there was no mutagenic potential observed for the damage induced by MBPDT, since the few cells that survived the treatment have lost their clonogenic capacity.

Conclusions

Our results elicit the potential of combined therapy in diminishing the toxicity of antineoplastic drugs. Ultimately, photodynamic therapy mediated by either methylene blue or Photogem as monotherapy or in combination with cisplatin has low mutagenic potential, which supports its safe use in clinical practice for the treatment of cervical cancer.
Literature
2.
go back to reference Brown SB, Brown EA, Walker I. Review the present and future role of photodynamic therapy in cancer treatment photodynamic therapy. Lancet Oncol. 2004;5:497–508.CrossRefPubMed Brown SB, Brown EA, Walker I. Review the present and future role of photodynamic therapy in cancer treatment photodynamic therapy. Lancet Oncol. 2004;5:497–508.CrossRefPubMed
3.
4.
go back to reference Zuluaga M-F, Lange N. Combination of photodynamic therapy with anti-cancer agents. Curr Med Chem. 2008;15(17):1655–73.CrossRefPubMed Zuluaga M-F, Lange N. Combination of photodynamic therapy with anti-cancer agents. Curr Med Chem. 2008;15(17):1655–73.CrossRefPubMed
5.
go back to reference de Freitas LM, Soares CP, Fontana CR. Synergistic effect of photodynamic therapy and cisplatin: A novel approach for cervical cancer. J Photochem Photobiol B. 2014;140C:365–73.CrossRef de Freitas LM, Soares CP, Fontana CR. Synergistic effect of photodynamic therapy and cisplatin: A novel approach for cervical cancer. J Photochem Photobiol B. 2014;140C:365–73.CrossRef
6.
go back to reference Kroemer G, et al. Classification of cell death: recommendations of the nomenclature committee on cell death 2009. Cell Death Differ. 2009;16:3–11.CrossRefPubMed Kroemer G, et al. Classification of cell death: recommendations of the nomenclature committee on cell death 2009. Cell Death Differ. 2009;16:3–11.CrossRefPubMed
7.
go back to reference Panzarini E, Inguscio V, Dini L. Immunogenic cell death: can it be exploited in PhotoDynamic therapy for cancer? BioMed Res Int. 2013;2013:4821–60.CrossRef Panzarini E, Inguscio V, Dini L. Immunogenic cell death: can it be exploited in PhotoDynamic therapy for cancer? BioMed Res Int. 2013;2013:4821–60.CrossRef
8.
go back to reference Gollnick SO. Photodynamic therapy and antitumor immunity. J Natl Compr Canc Netw. 2012;10:S-40–3. Gollnick SO. Photodynamic therapy and antitumor immunity. J Natl Compr Canc Netw. 2012;10:S-40–3.
9.
go back to reference Muller M, et al. One, two, three–p53, p63, p73 and chemosensitivity. Drug Resist Update. 2006;9:288–306.CrossRef Muller M, et al. One, two, three–p53, p63, p73 and chemosensitivity. Drug Resist Update. 2006;9:288–306.CrossRef
10.
go back to reference Coultas L, Strasser A. The role of the Bcl-2 protein family in cancer. Semin Cancer Biol. 2003;13:115–23.CrossRefPubMed Coultas L, Strasser A. The role of the Bcl-2 protein family in cancer. Semin Cancer Biol. 2003;13:115–23.CrossRefPubMed
11.
go back to reference Franken NA, et al. Clonogenic assay of cells in vitro. Nat Protocol. 2006;1(5):2315–9.CrossRef Franken NA, et al. Clonogenic assay of cells in vitro. Nat Protocol. 2006;1(5):2315–9.CrossRef
12.
go back to reference Blásquez-Castro A, et al. Differential photodynamic response of cultured cells to methylene blue and toluidine blue: role of dark redox processes. Photochem Photobiol Sci. 2009;8(3):371–6.CrossRef Blásquez-Castro A, et al. Differential photodynamic response of cultured cells to methylene blue and toluidine blue: role of dark redox processes. Photochem Photobiol Sci. 2009;8(3):371–6.CrossRef
13.
go back to reference Chen Y, et al. Apoptosis induced by methylene-blue-mediated photodynamic therapy in melanomas and the involvement of mitochondrial dysfunction revealed by proteomics. Cancer Sci. 2008;99(10):2019–27.PubMed Chen Y, et al. Apoptosis induced by methylene-blue-mediated photodynamic therapy in melanomas and the involvement of mitochondrial dysfunction revealed by proteomics. Cancer Sci. 2008;99(10):2019–27.PubMed
14.
go back to reference Tardivo J, et al. Methylene blue in photodynamic therapy: From basic mechanisms to clinical applications. Photodiagnosis Photodyn Ther. 2005;2(3):175–91.CrossRefPubMed Tardivo J, et al. Methylene blue in photodynamic therapy: From basic mechanisms to clinical applications. Photodiagnosis Photodyn Ther. 2005;2(3):175–91.CrossRefPubMed
16.
go back to reference Kim HS, Lee M-S. Essential Role of STAT1 in Caspase-Independent Cell Death of Activated Macrophages through the p38 Mitogen-Activated Protein Kinase/STAT1/Reactive Oxygen Species Pathway. Mol Cell Biol. 2005;25(15):6821–33.CrossRefPubMedPubMedCentral Kim HS, Lee M-S. Essential Role of STAT1 in Caspase-Independent Cell Death of Activated Macrophages through the p38 Mitogen-Activated Protein Kinase/STAT1/Reactive Oxygen Species Pathway. Mol Cell Biol. 2005;25(15):6821–33.CrossRefPubMedPubMedCentral
17.
go back to reference Lin Y, et al. Tumor necrosis factor-induced nonapoptotic cell death requires receptor-interacting protein-mediated cellular reactive oxygen species accumulation. J Biol Chem. 2004;279:10822–8.CrossRefPubMed Lin Y, et al. Tumor necrosis factor-induced nonapoptotic cell death requires receptor-interacting protein-mediated cellular reactive oxygen species accumulation. J Biol Chem. 2004;279:10822–8.CrossRefPubMed
18.
go back to reference Rogakou EP, et al. Initiation of DNA fragmentation during apoptosis induces phosphorylation of H2AX histone at serine 139. J Biol Chem. 2000;275(13):9390–5.CrossRefPubMed Rogakou EP, et al. Initiation of DNA fragmentation during apoptosis induces phosphorylation of H2AX histone at serine 139. J Biol Chem. 2000;275(13):9390–5.CrossRefPubMed
19.
20.
22.
go back to reference Kamat JP, Devasagayam TP. Methylene blue plus light-induced lipid peroxidation in rat liver microsomes: inhibition by nicotinamide (vitamin B3) and other antioxidants. Chem Biol Interact. 1996;99(1-3):1–16.CrossRefPubMed Kamat JP, Devasagayam TP. Methylene blue plus light-induced lipid peroxidation in rat liver microsomes: inhibition by nicotinamide (vitamin B3) and other antioxidants. Chem Biol Interact. 1996;99(1-3):1–16.CrossRefPubMed
23.
go back to reference Rello S, et al. Morphological criteria to distinguish cell death induced by apoptotic and necrotic treatments. Apoptosis. 2005;10(1):201–8.CrossRefPubMed Rello S, et al. Morphological criteria to distinguish cell death induced by apoptotic and necrotic treatments. Apoptosis. 2005;10(1):201–8.CrossRefPubMed
24.
go back to reference Hsieh Y-J, et al. Subcellular localization of Photofrin determines the death phenotype of human epidermoid carcinoma A431 cells triggered by photodynamic therapy: when plasma membranes are the main targets. J Cell Physio. 2003;194(3):363–75.CrossRef Hsieh Y-J, et al. Subcellular localization of Photofrin determines the death phenotype of human epidermoid carcinoma A431 cells triggered by photodynamic therapy: when plasma membranes are the main targets. J Cell Physio. 2003;194(3):363–75.CrossRef
25.
go back to reference Dellinger M. Apoptosis or necrosis following Photofrin photosensitization: influence of the incubation protocol. Photochem Photobiol. 1996;64:182–7.CrossRefPubMed Dellinger M. Apoptosis or necrosis following Photofrin photosensitization: influence of the incubation protocol. Photochem Photobiol. 1996;64:182–7.CrossRefPubMed
26.
27.
go back to reference Lüthi AU, Martin SJ. The CASBAH: a searchable database of caspase substrates. Cell Death Differ. 2007;14:641–50.CrossRefPubMed Lüthi AU, Martin SJ. The CASBAH: a searchable database of caspase substrates. Cell Death Differ. 2007;14:641–50.CrossRefPubMed
29.
go back to reference Porter AG, Jänicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 1999;6(2):99–104.CrossRefPubMed Porter AG, Jänicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ. 1999;6(2):99–104.CrossRefPubMed
30.
go back to reference Bro LE, Kruyt FAE, Giaccone G. Cell death independent of caspases : a review. Clin Cancer Res. 2005;11(9):3155–63.CrossRef Bro LE, Kruyt FAE, Giaccone G. Cell death independent of caspases : a review. Clin Cancer Res. 2005;11(9):3155–63.CrossRef
31.
go back to reference Ekert PG, et al. Apaf-1 and caspase-9 accelerate apoptosis, but do not determine whether factor-deprived or drug-treated cells die. J Cell Biol. 2004;165:835–42.CrossRefPubMedPubMedCentral Ekert PG, et al. Apaf-1 and caspase-9 accelerate apoptosis, but do not determine whether factor-deprived or drug-treated cells die. J Cell Biol. 2004;165:835–42.CrossRefPubMedPubMedCentral
32.
go back to reference Tesniere A, et al. Molecular characteristics of immunogenic cancer cell death. Cell Death Differ. 2008;15(1):3–12.CrossRefPubMed Tesniere A, et al. Molecular characteristics of immunogenic cancer cell death. Cell Death Differ. 2008;15(1):3–12.CrossRefPubMed
33.
go back to reference Melcher A, et al. Tumor immunogenicity is determined by the mechanism of cell death via induction of heat shock protein expression. Nat Med. 1998;4(5):581–87.CrossRefPubMed Melcher A, et al. Tumor immunogenicity is determined by the mechanism of cell death via induction of heat shock protein expression. Nat Med. 1998;4(5):581–87.CrossRefPubMed
34.
go back to reference Goldszmid RS, et al. Dendritic cells charged with apoptotic tumor cells induce long-lived protective CD4 + and CD8 + T cell immunity against B16 melanoma. J Immunol. 2003;171(11):5940–7.CrossRefPubMed Goldszmid RS, et al. Dendritic cells charged with apoptotic tumor cells induce long-lived protective CD4 + and CD8 + T cell immunity against B16 melanoma. J Immunol. 2003;171(11):5940–7.CrossRefPubMed
35.
go back to reference Scheffer SRS, et al. Apoptotic, but not necrotic, tumor cell vaccines induce a potent immune response in vivo. Int J Cancer. 2003;211(2002):205–11.CrossRef Scheffer SRS, et al. Apoptotic, but not necrotic, tumor cell vaccines induce a potent immune response in vivo. Int J Cancer. 2003;211(2002):205–11.CrossRef
36.
go back to reference Larmonier N, et al. An atypical caspase-independent death pathway for an immunogenic cancer cell line. Oncogene. 2002;21(39):6091–100.CrossRefPubMed Larmonier N, et al. An atypical caspase-independent death pathway for an immunogenic cancer cell line. Oncogene. 2002;21(39):6091–100.CrossRefPubMed
37.
go back to reference Davies J, et al. Methylene blue but not indigo carmine causes DNA damage to colonocytes in vitro and in vivo at concentrations used in clinical chromoendoscopy. Gut. 2007;56(1):155–6.CrossRefPubMedPubMedCentral Davies J, et al. Methylene blue but not indigo carmine causes DNA damage to colonocytes in vitro and in vivo at concentrations used in clinical chromoendoscopy. Gut. 2007;56(1):155–6.CrossRefPubMedPubMedCentral
38.
go back to reference Woods JA, et al. The effect of photofrin on DNA strand breaks and base oxidation in HaCaT keratinocytes: a comet assay study. Photochem Photobiol. 2004;79(1):105–13.CrossRefPubMed Woods JA, et al. The effect of photofrin on DNA strand breaks and base oxidation in HaCaT keratinocytes: a comet assay study. Photochem Photobiol. 2004;79(1):105–13.CrossRefPubMed
39.
go back to reference Epe B, Pflaum M, Boiteux S. DNA damage induced by photosensitizers in cellular and cell-free systems. Mutat Res. 1993;299(3–4):135–45.CrossRefPubMed Epe B, Pflaum M, Boiteux S. DNA damage induced by photosensitizers in cellular and cell-free systems. Mutat Res. 1993;299(3–4):135–45.CrossRefPubMed
40.
41.
go back to reference Cipriani G, et al. Nuclear poly(ADP-ribose) polymerase-1 rapidly triggers mitochondrial dysfunction. J Biol Chem. 2005;280(17):17227–34.CrossRefPubMed Cipriani G, et al. Nuclear poly(ADP-ribose) polymerase-1 rapidly triggers mitochondrial dysfunction. J Biol Chem. 2005;280(17):17227–34.CrossRefPubMed
42.
43.
go back to reference Alano CC, Ying W, Swanson RA. Poly(ADP-ribose) polymerase-1-mediated cell death in astrocytes requires NAD+ depletion and mitochondrial permeability transition. J Biol Chem. 2004;279:18895–902.CrossRefPubMed Alano CC, Ying W, Swanson RA. Poly(ADP-ribose) polymerase-1-mediated cell death in astrocytes requires NAD+ depletion and mitochondrial permeability transition. J Biol Chem. 2004;279:18895–902.CrossRefPubMed
44.
go back to reference Burkart V, et al. Mice lacking the poly(ADP-ribose) polymerase gene are resistant to pancreatic beta-cell destruction and diabetes development induced by streptozocin. Nat Med. 1999;5:314–9.CrossRefPubMed Burkart V, et al. Mice lacking the poly(ADP-ribose) polymerase gene are resistant to pancreatic beta-cell destruction and diabetes development induced by streptozocin. Nat Med. 1999;5:314–9.CrossRefPubMed
45.
go back to reference Eliasson MJ, et al. Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med. 1997;3:1089–95.CrossRefPubMed Eliasson MJ, et al. Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med. 1997;3:1089–95.CrossRefPubMed
46.
go back to reference Cosi C, et al. Poly(ADP-ribose) polymerase: early involvement in glutamate-induced neurotoxicity in cultured cerebellar granule cells. J Neurosci Res. 1994;39:38–46.CrossRefPubMed Cosi C, et al. Poly(ADP-ribose) polymerase: early involvement in glutamate-induced neurotoxicity in cultured cerebellar granule cells. J Neurosci Res. 1994;39:38–46.CrossRefPubMed
47.
go back to reference Mcnair FI, et al. A comet assay of DNA damage and repair in K562 cells after photodynamic therapy using haematoporphyrin derivative, methylene blue and meso-tetrahydroxyphenylchlorin. Br J Cancer. 1997;75(12):1721–9.CrossRefPubMedPubMedCentral Mcnair FI, et al. A comet assay of DNA damage and repair in K562 cells after photodynamic therapy using haematoporphyrin derivative, methylene blue and meso-tetrahydroxyphenylchlorin. Br J Cancer. 1997;75(12):1721–9.CrossRefPubMedPubMedCentral
49.
go back to reference Shishkova N, Kuznetsova O, Berezov T. Photodynamic therapy for gynecological diseases and breast cancer. Cancer Biol Med. 2012;9(1):9–17.PubMedPubMedCentral Shishkova N, Kuznetsova O, Berezov T. Photodynamic therapy for gynecological diseases and breast cancer. Cancer Biol Med. 2012;9(1):9–17.PubMedPubMedCentral
50.
go back to reference Sakamoto M. Safety guidelines for photodynamic therapy in the treatment of early stage cancer and dysplasia of the uterine cervix. Laser Ther. 2012;21(1):60–4.CrossRefPubMedPubMedCentral Sakamoto M. Safety guidelines for photodynamic therapy in the treatment of early stage cancer and dysplasia of the uterine cervix. Laser Ther. 2012;21(1):60–4.CrossRefPubMedPubMedCentral
51.
go back to reference Yamaguchi S, et al. Photodynamic therapy for cervical intraepithelial neoplasia. Oncology. 2005;69:110–6.CrossRefPubMed Yamaguchi S, et al. Photodynamic therapy for cervical intraepithelial neoplasia. Oncology. 2005;69:110–6.CrossRefPubMed
52.
go back to reference Muroya T, et al. Phase III clinical trial of photodynamic therapy for early cancer and pre-cancerous lesion of the uterine cervix using porfimer sodium (PHE) and excimer dye laser (PDT EDL-1). Oncol Chemother. 1992;8:302–7. Muroya T, et al. Phase III clinical trial of photodynamic therapy for early cancer and pre-cancerous lesion of the uterine cervix using porfimer sodium (PHE) and excimer dye laser (PDT EDL-1). Oncol Chemother. 1992;8:302–7.
Metadata
Title
Photodynamic therapy combined to cisplatin potentiates cell death responses of cervical cancer cells
Authors
Laura Marise de Freitas
Rodolfo Bortolozo Serafim
Juliana Ferreira de Sousa
Thaís Fernanda Moreira
Cláudia Tavares dos Santos
Amanda Martins Baviera
Valeria Valente
Christiane Pienna Soares
Carla Raquel Fontana
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2017
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-017-3075-1

Other articles of this Issue 1/2017

BMC Cancer 1/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine