HCT116 Kolorektal Kanser Hücre Sağ-kalımının Paklitaksel ve Olaparib Kombinasyon Tedavisiyle Engellenmesi

Kolorektal kanser, dünya genelinde kansere bağlı ölümün temel sebeplerinden biridir ve konvansiyonel kemoterapi yaklaşımları kolorektal kanser tedavisinin klinik yönetimine öncülük etmektedir. Paklitaksel dahil olmak üzere çoğu kemoterapötik, kolorektal kanser hastalarını tedavi etmede etkili bir şekilde kullanılmasına rağmen, bazı tümör hücreleri özellikle çeşitli DNA tamir aktivitelerine bağımlılık kazanmak suretiyle çok sayıda hücresel mekanizmaların düzenlenmesini arttırarak ilaç direnci kazanırlar. Çeşitli DNA hasar yanıtı bileşenlerini hedefleyen küçük-moleküllü inhibitörlerin klinik öncesi değerlendirmesi günümüzde kanser araştırmalarının aktif bir alanını oluşturmaktadır. Bu çalışma, HCT116 kolorektal kanser hücrelerinde paklitaksel ile kombine edilen küçük-moleküllü PARP inhibitörü olaparibin sitotoksik etkisini değerlendirmeyi amaçlamıştır. Gerçekleştirdiğimiz koloni sağ-kalım analizleri paklitaksel ve olaparib kombinasyonunun sadece paklitaksel ya da olaparib tedavisi ile kıyas edildiğinde kolorektal kanser hücrelerinin sağ kalımını önemli derecede inhibe ettiğini gösterdi. Western blot ve immünofloresan olmak üzere iki farklı immünolojik yöntem gerçekleştirilerek, olaparibin paklitaksel tedavisinin oluşturduğu DNA hasarı birikimini yoğunlaştırdığı tespit edildi. Sonuç olarak, HCT116 kolorektal kanser hücreleriyle gerçekleştirdiğimiz klinik öncesi çalışmamız, olaparib ve paklitaksel kombinasyonunun kolorektal kanser tedavisi için potansiyel bir tedavi yaklaşımı sunabileceğini göstermektedir.

Inhibition of Colorectal Cancer Cell Survival by Paclitaxel Combined with Olaparib

Colorectal cancer is one of the main reasons for cancer-related mortality around the world and conventional chemotherapy approaches lead the clinical management of colorectal cancer treatment. Although many chemotherapeutics including paclitaxel have been effectively used to treat colorectal cancer patients, some tumour cells acquire drug resistance by increasing the regulation of various cellular mechanisms, primarily becoming addictive to some DNA repair activities. Preclinical evaluation of small-molecule inhibitors targeting various DNA damage response components is currently an active area of cancer research. This study aimed to assess the cytotoxic effect of paclitaxel combined with small-molecule PARP inhibitor olaparib in HCT116 colorectal cancer cells. We conducted clonogenic assays, showing that the paclitaxel and olaparib combination significantly inhibits the survival of colorectal cancer cells compared to paclitaxel or olaparib alone. By performing two distinct immunological methodology, Western blotting and immunofluorescence, we further demonstrated that olaparib intensifies the DNA damage accumulation induced by paclitaxel treatment. In conclusion, our preclinical study with HCT116 colorectal cancer cells suggests that olaparib may potentially serve as a combination partner to paclitaxel for effective colorectal cancer treatment.

___

  • [1] Bray, F., Ferlay, J., Soerjomataram, I., Siegel, R.L., Torre, L.A., Jemal, A., Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries, CA: A Cancer Journal for Clinicians, 68(6), 394-424, 2018.
  • [2] Ades, S., Adjuvant chemotherapy for colon cancer in the elderly: Moving from evidence to practice, Oncology, 23(2), 162-167, 2009.
  • [3] Jensen, N.F., et al., Establishment and characterization of models of chemotherapy resistance in colorectal cancer: Towards a predictive signature of chemoresistance, Molecular Oncology, 9(6), 1169-1185, 2015.
  • [4] Othman, T., Goto, S., Lee, J.B., Taimura, A., Matsumoto, T., Kosaka, M., Hyperthermic enhancement of the apoptotic and antiproliferative activities of paclitaxel, Pharmacology, 62(4), 208-212, 2001.
  • [5] Barbuti, A.M., Chen, Z.S., Paclitaxel through the ages of anticancer therapy: Exploring its role in chemoresistance and radiation therapy, Cancers, 7(4), 2360-2371, 2015.
  • [6] Rowinsky, E.K., Donehower, R.C., Paclitaxel (Taxol), New England Journal of Medicine, 332(15), 1004-1014, 1995.
  • [7] Banerji, A., Lax, T., Guyer, A., Hurwitz, S., Camargo, C.A., Long, A.A., Management of hypersensitivity reactions to carboplatin and paclitaxel in an outpatient oncology infusion center: A 5-year review, The Journal of Allergy and Clinical Immunology: In Practice, 2(4), 428- 433, 2014.
  • [8] Orr, G.A., Verdier-Pinard, P., McDaid, H., Horwitz, S.B., Mechanisms of Taxol resistance related to microtubules, Oncogene, 22(47), 7280-7295, 2003.
  • [9] Lien, K., Georgsdottir, S., Sivanathan, L., Chan, K., Emmenegger, U., Low-dose metronomic chemotherapy: A systematic literature analysis, Eurepean Journal of Cancer, 49(16), 3387-3395, 2013.
  • [10] Loven, D., Hasnis, E., Bertolini, F., Shaked, Y., Low-dose metronomic chemotherapy: From past experience to new paradigms in the treatment of cancer, Drug Discovery Today, 18(3- 4), 193-201, 2013.
  • [11] Gong, L.H., et al., Piperlongumine induces apoptosis and synergizes with cisplatin or paclitaxel in human ovarian cancer cells, Oxidative Medicine and Cellular Longevity, 906804, 1-10, 2014.
  • [12] Lv, C., et al., Low-dose paclitaxel inhibits tumor cell growth by regulating glutaminolysis in colorectal carcinoma cells, Frontiers in Pharmacology, 8(244), 1-10, 2017.
  • [13] Cheng, M., et al., Weekly dose-dense paclitaxel and triweekly low-dose cisplatin: A well-tolerated and effective chemotherapeutic regimen for first-line treatment of advanced ovarian, fallopian tube, and primary peritoneal cancer, International Journal of Environmental Research and Public Health, 16(23), 4794, 2019.
  • [14] Kennedy, A.S., Harrison, G. H., Mansfield, C. M., Zhou, X. J., Xu, J. F., Balcer‐ Kubiczek, E. K., Survival of colorectal cancer cell lines treated with paclitaxel, radiation, and 5‐ FU: Effect of TP53 or hMLH1 deficiency, International Journal of Cancer, 90(4), 175-185, 2000.
  • [15] Li, H., et al., Low-dose paclitaxel downregulates MYC proto-oncogene bHLH transcription factor expression in colorectal carcinoma cells, Oncology Letters, 15(2), 1881- 1887, 2018.
  • [16] Li, Q., et al., Low doses of paclitaxel enhance liver metastasis of breast cancer cells in the mouse model, The FEBS Journal, 283(15), 2836-2852, 2016.
  • [17] Shetti, D., Zhang, B., Fan, C., Mo, C., Lee, B.H., Wei, K., Low Dose of Paclitaxel Combined with XAV939 Attenuates Metastasis, Angiogenesis and Growth in Breast Cancer by Suppressing Wnt Signaling, Cells, 8(8), 1-22, 2019.
  • [18] Ciccia, A., Elledge, S.J., The DNA Damage Response: Making It Safe to Play with Knives, Molecular Cell, 40(2), 179-204, 2010.
  • [19] Goldstein, M., Kastan, M.B., The DNA Damage Response: Implications for Tumor Responses to Radiation and Chemotherapy, Annual Review of Medicine, 66(1), 129-143, 2015.
  • [20] Sonnenblick, A., de Azambuja, E., Azim, H.A., Piccart, M., An update on PARP inhibitors-moving to the adjuvant setting, Nature Review Clinical Oncology, 12(1), 27-41, 2015.
  • [21] Morales, J.C., et al., Review of poly (ADP-ribose) polymerase (PARP) mechanisms of action and rationale for targeting in cancer and other diseases, Critical Reviews in Eukaryotic Gene Expression., 24(1), 15-28, 2014.
  • [22] Dréan, A., Lord, C.J., Ashworth, A., PARP inhibitor combination therapy, Critical Reviews in Oncology/Hematology, 108, 73-85, 2016.
  • [23] Walsh, C., Targeted therapy for ovarian cancer: The rapidly evolving landscape of PARP inhibitor use, Minerva Ginecologica, 70(2), 150-170, 2018.
  • [24] The Food and Drug Administration (FDA). (2020, May 01). FDA approves olaparib for germline BRCA-mutated metastatic breast cancer, 2018.
  • [25] The Food and Drug Administration (FDA). (2020, May 01). FDA approves olaparib for gBRCAm metastatic pancreatic adenocarcinoma, 2019.
  • [26] Arena, S., et al., A subset of colorectal cancers with cross-sensitivity to olaparib and oxaliplatin, Clinical Cancer Research, 26(6), 1372-1384, 2020.
  • [27] Augustine, T., Maitra, R., Zhang, J., Nayak, J., Goel, S., Sensitization of colorectal cancer to irinotecan therapy by PARP inhibitor rucaparib, Investigational New Drugs, 37(5), 948-960, 2019.
  • [28] Genther Williams S.M., et al., Treatment with the PARP inhibitor, niraparib, sensitizes colorectal cancer cell lines to irinotecan regardless of MSI/MSS status, Cancer Cell International., 15(1), 1-11, 2015.
  • [29] Gomez, V., et al., Regulation of DNA damage responses and cell cycle progression by hMOB2, Cellular Signalling., 27(2), 326–39, 2015.
  • [30] Hergovich, A., Bichsel, S.J., Hemmings, B.A., Human NDR kinases are rapidly activated by MOB proteins through recruitment to the plasma membrane and phosphorylation, Molecular and Cellular Biology, 25(18), 8259-8272, 2005.
  • [31] Weaver, B.A., How Taxol/paclitaxel kills cancer cells, Molecular Biology of the Cell, 25(18), 2677-2681, 2014.
  • [32] Berg, K.C.G., et al., Multi-omics of 34 colorectal cancer cell lines - a resource for biomedical studies, Molecular Cancer, 16(116), 1-16, 2017.
  • [33] Duldulao, M.P., et al., Gene expression variations in microsatellite stable and unstable colon cancer cells, Journal of Surgical Research, 174(1), 1-6, 2012.
  • [34] Shen, J., et al., ARID1A Deficiency Impairs the DNA Damage Checkpoint and Sensitizes Cells to PARP Inhibitors, Cancer Discovery, 5(7), 752-767, 2015.
  • [35] Wang, C., Jette, N., Moussienko, D., Gwyn Bebb, D., Lees-Miller, S.P., ATMDeficient Colorectal Cancer Cells Are Sensitive to the PARP Inhibitor Olaparib, Translational. Oncology, 10(2), 190-196, 2017.
  • [36] Kuo, L.J., Yang, L.X., γ-H2AX- A novel biomaker for DNA double-strand breaks, In Vivo, 22(3), 305-309, 2008.
  • [37] Desai, A., Yan, Y., Gerson, S.L., Advances in therapeutic targeting of the DNA damage response in cancer, DNA Repair, 66(67), 24-29, 2018.
  • [38] Masutani, M., Nozaki, T., Wakabayashi, K., Sugimura, T., Role of poly(ADP-ribose) polymerase in cell-cycle checkpoint mechanisms following γ-irradiation, Biochimie, 77(6), 462- 465, 1995.
  • [39] Ollikainen, T., et al., In vitro sensitivity of normal human mesothelial and malignant mesothelioma cell lines to four new chemotherapeutic agents, Anticancer Drugs, 11(2), 93-99, 2000.
  • [40] Digue, L., et al., Evaluation of the genotoxic activity of paclitaxel by the in vitro micronucleus test in combination with fluorescent in situ hybridization of a DNA centromeric probe and the alkaline single cell gel electrophoresis technique (comet assay) in human T-lymph, Environmental and Molecular Mutagen, 34(4), 269-278, 1999.
  • [41] Branham, M.T., Nadin, S.B., Vargas-Roig, L.M., Ciocca, D.R., DNA damage induced by paclitaxel and DNA repair capability of peripheral blood lymphocytes as evaluated by the alkaline comet assay, Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 560(1), 11-17, 2004.
  • [42] Bryant, H.E., Helleday, T., Inhibition of poly (ADP-ribose) polymerase activates ATM which is required for subsequent homologous recombination repair, Nucleic Acids Research, 34(6), 1685-1691, 2006.