AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (4.6 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Efficacy of the new therapeutic approach in curing malignant neoplasms on the model of human glioblastoma

Evgeniya V. Dolgova1Oleg M. Andrushkevich2Polina E. Kisaretova1Anastasia S. Proskurina1Genrikh S. Ritter1Tatyana D. Dubatolova3Margarita V. Romanenko4Oleg S. Taranov5Yaroslav R. Efremov1,4Evgeniy L. Zavyalov1Alexandr V. Romaschenko1Sergey V. Mishinov6Svetlana S. Kirikovich1Evgeniy V. Levites1Ekaterina A. Potter1Alexandr A. Ostanin7Elena R. Chernykh7Stanislav Yu. Roshchin8Anatoliy V. Bervitskiy9Galina I. Moysak4,9Jamil A. Rzaev4,9Sergey S. Bogachev1 ( )
Institute of Cytology and Genetics SB RAS, Novosibirsk 630090, Russia
A.I. Evdokimov Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
Institute of Molecular and Cellular Biology, Novosibirsk 630090, Russia
Novosibirsk State University, Novosibirsk 630090, Russia
The State Research Center of Virology and Biotechnology “Vector”, Koltsovo, Novosibirsk 630559, Russia
First Department of Neurosurgery, Ya. L. Tsivian Novosibirsk Research Institute of Traumatology and Orthopaedics, Novosibirsk 630091, Russia
Institute of Fundamental and Clinical immunology, Novosibirsk 630099, Russia
Sklifosovsky Research Institute of Emergency Medicine, Moscow 129010, Russia
Federal Center of Neurosurgery, Novosibirsk 630048, Russia
Show Author Information

Abstract

Objective

Glioma is a highly invasive tumor, frequently disposed in essential areas of the brain, which makes its surgical excision extremely difficult; meanwhile adjuvant therapy remains quite ineffective.

Methods

In the current report, a new therapeutic approach in curing malignant neoplasms has been performed on the U87 human glioblastoma model. This approach, termed “Karanahan”, is aimed at the eradication of cancer stem cells (CSCs), which were recently shown to be capable of internalizing fragments of extracellular double-stranded DNA. After being internalized, these fragments interfere in the process of repairing interstrand cross-links caused by exposure to appropriate cytostatics, and such an interference results either in elimination of CSCs or in the loss of their tumorigenic potency. Implementation of the approach requires a scheduled administration of cytostatic and complex composite double-stranded DNA preparation.

Results

U87 cells treated in vitro in accordance with the Karanahan approach completely lost their tumorigenicity and produced no grafts upon intracerebral transplantation into immunodeficient mice. In SCID mice with developed subcutaneous grafts, the treatment resulted in reliable slowing down of tumor growth rate (P < 0.05). In the experiment with intracerebral transplantation of U87 cells followed by surgical excision of the developed graft and subsequent therapeutic treatment, the Karanahan approach was shown to reliably slow down the tumor growth rate and increase the median survival of the mice twofold relative to the control.

Conclusions

The effectiveness of the Karanahan approach has been demonstrated both in vitro and in vivo in treating developed subcutaneous grafts as well as orthotopic grafts after surgical excision of the tumor.

Electronic Supplementary Material

Download File(s)
cbm-18-3-910-ESM.pdf (5.2 MB)

References

1

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005; 352: 987-96.

2

Konar SK, Bir SC, Maiti TK, Nanda A. A systematic review of overall survival in pediatric primary glioblastoma multiforme of the spinal cord. J Neurosurg Pediatr. 2017; 19: 239-48.

3

Smith MA, Freidlin B, Gloeckler Ries LA, Simon R. Trends in reported incidence of primary malignant brain tumors in children in the United States. JNCI J Natl Cancer Inst. 1998; 90: 1269-77.

4

McNeill KA. Epidemiology of brain tumors. Neurol Clin. 2016; 34: 981-98.

5
Gilbert CA, Ross AH. Glioma stem cells: cell culture, markers and targets for new combination therapies. In: Shostak S, editor. Cancer Stem Cells Theor Pract. London, UK: IntechOpen, 2011. p. 80-104.
6

Friedman HS, Kerby T. Temozolomide and treatment of malignant glioma 1. Clin Cancer Res. 2000; 6: 2585-97.

7

Ramirez YP, Weatherbee JL, Wheelhouse RT, Ross AH. Glioblastoma multiforme therapy and mechanisms of resistance. Pharmaceuticals. 2013; 6: 1475-506.

8

Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V, et al. Inactivation of the DNA-repair gene MGMT and clinical response of gliomas to alkylating agents. N Engl J Med. 2000; 343: 1350-4.

9

Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-Cortes J, et al. A cell initiating human acute myeloid leukamia after transplantation into SCID mice. Nature. 1994; 367: 645-8.

10

Bonnet D, Dick JE. Human acute myeloid leukemia is organized as hierarchy that originates from a primitive hematopoietic cell. Nat Med. 1997; 3: 730-7.

11

Dolgova EV, Mishinov SV, Proskurina AS, Potter EA, Efremov YR, Bayborodin SI, et al. Novel cancer stem marker and its applicability for grading primary human gliomas. Technol Cancer Res Treat. 2018; 17: 1-8.

12

Brescia P, Richichi C, Pelicci G. Current strategies for identification of glioma stem cells: adequate or unsatisfactory? J Oncol. 2012; 2012: 376894.

13

Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006; 444: 756-60.

14

Wu A, Wei J, Kong L-Y, Wang Y, Priebe W, Qiao W, et al. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro Oncol. 2010; 12: 1113-25.

15

Jinesh GG, Choi W, Shah JB, Lee EK, Willis DL, Kamat AM. Blebbishields, the emergency program for cancer stem cells: sphere formation and tumorigenesis after apoptosis. Cell Death Differ. 2013; 20: 382-95.

16

Efremov YR, Proskurina AS, Potter EA, Dolgova EV, Efremova OV, Taranov OS, et al. Cancer stem cells: emergent nature of tumor emergency. Front Genet. 2018; 9: 544.

17

Chen J, Kesari S, Rooney C, Strack PR, Chen J, Shen H, et al. Inhibition of Notch signaling blocks growth of glioblastoma cell lines and tumor neurospheres. Genes and Cancer. 2010; 1: 822-35.

18

Fan X, Khaki L, Zhu TS, Soules ME, Talsma CE, Gul N, et al. NOTCH pathway blockade depletes CD133-positive glioblastoma. Stem Cells. 2010; 28: 5-16.

19

Liu Y, Liu X, Chen L, Du W-Z, Cui Y-Q, Piao X-Y, et al. Targeting glioma stem cells via the Hedgehog signaling pathway. Neuroimmunol Neuroinflammation. 2014; 1: 51-9.

20

Takezaki T, Hide T, Takanaga H, Nakamura H, Kuratsu J-I, Kondo T. Essential role of the Hedgehog signaling pathway in human glioma-initiating cells. Cancer Sci. 2011; 102: 1306-12.

21

Wei K-C, Huang C-Y, Chen P-Y, Feng L-Y, Wu T-WE, Chen S-M, et al. Evaluation of the prognostic value of CD44 in glioblastoma multiforme. Anticancer Res. 2010; 30: 253-9.

22

Kase M, Minajeva A, Niinepuu K, Kase S, Vardja M, Asser T, et al. Impact of CD133 positive stem cell proportion on survival in patients with glioblastoma multiforme. Radiol Oncol. 2013; 47: 405-10.

23

Brescia P, Ortensi B, Fornasari L, Levi D, Broggi G, Pelicci G. CD133 is essential for glioblastoma stem cell maintenance. Stem Cells. 2013; 31: 857-69.

24

Pietras A, Katz AM, Ekström EJ, Wee B, Halliday JJ, Pitter KL, et al. Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth. Cell Stem Cell. 2014; 14: 357-69.

25

Dolgova EV, Efremov YR, Orishchenko KE, Andrushkevich OM, Alyamkina EA, Proskurina AS, et al. Delivery and processing of exogenous double-stranded DNA in mouse CD34+ hematopoietic progenitor cells and their cell cycle changes upon combined treatment with cyclophosphamide and double-stranded DNA. Gene. 2013; 528: 74-83.

26

Dolgova EV, Alyamkina EA, Efremov YR, Nikolin VP, Popova NA, Tyrinova TV, et al. Identification of cancer stem cells and a strategy for their elimination. Cancer Biol Ther. 2014; 15: 1378-94.

27

Alyamkina EA, Nikolin VP, Popova NA, Minkevich AM, Kozel AV, Dolgova EV, et al. Combination of cyclophosphamide and double-stranded DNA demonstrates synergistic toxicity against established xenografts. Cancer Cell Int. 2015; 15: 32.

28

Potter EA, Dolgova EV, Proskurina AS, Minkevich AM, Efremov YR, Taranov OS, et al. A strategy to eradicate well-developed Krebs-2 ascites in mice. Oncotarget. 2016; 7: 11580-94.

29

Potter EA, Proskurina AS, Ritter GS, Dolgova EV, Nikolin VP, Popova NA, et al. Efficacy of a new cancer treatment strategy based on eradication of tumor-initiating stem cells in a mouse model of Krebs-2 solid adenocarcinoma. Oncotarget. 2018; 9: 28486-99.

30

Dolgova EV, Proskurina AS, Nikolin VP, Popova NA, Alyamkina EA, Orishchenko KE, et al. “Delayed death” phenomenon: a synergistic action of cyclophosphamide and exogenous DNA. Gene. 2012; 495: 134-45.

31

Kisaretova PE, Kirikovich SS, Ritter GS, Efremov YR, Taranov OS, Dubatolova TD, et al. Approbation of the cancer treatment approach based on the eradication of TAMRA+ cancer stem cells in a model of murine cyclophosphamide resistant lymphosarcoma. Anticancer Res. 2020; 40: 795-805.

32

Faustino-Rocha A, Oliveira PA, Pinho-Oliveira J, Teixeira-Guedes C, Soares-Maia R, da Costa RG, et al. Estimation of rat mammary tumor volume using caliper and ultrasonography measurements. Lab Anim (NY). 2013; 42: 217-24.

33

Dolgova EV, Proskurina AS, Potter EA, Tyrinova TV, Taranov OS, Efremov YR, et al. Evaluation of a strategy for tumor-initiating stem cell eradication in primary human glioblastoma cultures as a model. Vavilov J Genet Breed. 2018; 22: 825-36[in Russian].

34

Potter EA, Dolgova EV, Proskurina AS, Efremov YR, Minkevich AM, Rozanov AS, et al. Gene expression profiling of tumor-initiating stem cells from mouse Krebs-2 carcinoma using a novel marker of poorly differentiated cells. Oncotarget. 2017; 8: 9425-41.

35

Dolgova EV, Petrova DD, Proskurina AS, Ritter GS, Kisaretova PE, Potter EA, et al. Identification of the xenograft and its ascendant sphere-forming cell line as belonging to EBV-induced lymphoma, and characterization of the status of sphere-forming cells. Cancer Cell Int. 2019; 19: 1-21.

36

Karle P, Renner M, Salmons B, Günzburg WH. Necrotic, rather than apoptotic, cell death caused by cytochrome P450-activated ifosfamide. Cancer Gene Ther. 2001; 8: 220-30.

37

Hanada K, Budzowska M, Modesti M, Maas A, Wyman C, Essers J, et al. The structure-specific endonuclease Mus81-Eme1 promotes conversion of interstrand DNA crosslinks into double-strands breaks. EMBO J. 2006; 25: 4921-32.

38

Zhang N, Liu X, Li L, Legerski R. Double-strand breaks induce homologous recombinational repair of interstrand cross-links via cooperation of MSH2, ERCC1-XPF, REV3, and the Fanconi anemia pathway. DNA Repair (Amst). 2007; 6: 1670-8.

39

Collins AR. The comet assay for DNA damage and repair: principles, applications, and limitations. Mol Biotechnol. 2004; 26: 249-61.

40

Niedernhofer LJ, Odijk H, Budzowska M, van Drunen E, Maas A, Theil AF, et al. The structure-specific endonuclease Ercc1-Xpf is required to resolve DNA interstrand cross-link-induced double-strand breaks. Mol Cell Biol. 2004; 24: 5776-87.

41

Genka S, Deutsch J, Stahle PL, Shetty UH, John V, Robinson C, et al. Brain and plasma pharmacokinetics and anticancer activities of cyclophosphamide and phosphoramide mustard in the rat. Cancer Chemother Pharmacol. 1990; 27: 1-7.

42

Song X, Dilly A-K, Kim S-Y, Choudry H, Lee Y. Rapamycin-enhanced mitomycin C-induced apoptotic death is mediated through the S6K1–Bad–Bak pathway in peritoneal carcinomatosis. Cell Death Dis. 2014; 5: e1281.

43

Molyneux G, Gibson FM, Gordon-Smith EC, Pilling AM, Liu KC, Rizzo S, et al. The haemotoxicity of mitomycin in a repeat dose study in the female CD-1 mouse. Int J Exp Path. 2005; 86: 415-30.

44

Sosedova LM, Kudayeva Ⅳ, Titov EA, Yakimova NL, Masnavieva LB, Katamanova EV. Morphologic and neurochemical effects in long-term period of mercurial intoxication (experimental data). Russ J Occup Heal Ind Ecol. 2009; 1: 37-42[in Russian].

45

Dolgova EV, Efremov YR, Taranov OS, Potter EP, Nikolin VP, Popova NA, et al. Comparative analysis of pathologic processes developing in mice housed in SPF vs. non-SPF conditions and treated with cyclophosphamide and dsDNA preparation. Pathol Res Pract. 2015; 211: 754-8.

46

Leonova ZA. Priones and prion diseases. Acta Biomed Sci. 2010; 6: 169-74.

47

Boychenko MN, Volchkova YV, Pak SG. Biological Preparations and prion diseases: whether etioligical kelation is possible? Int Neurol J. 2014; 6: 77-84.

48

Dolgova EV, Shevela EY, Tyrinova TV, Minkevich AM, Proskurina AS, Potter EA, et al. Nonadherent spheres with multiple myeloma surface markers contain cells that contribute to sphere formation and are capable of internalizing extracellular double-stranded DNA. Clin Lymphoma Myeloma Leuk. 2016; 16: 563-76.

49

Potter EA, Dolgova EV, Minkevich AM, Proskurina AS, Efremov YR, Taranov OS, et al. Analysis of different therapeutic schemes combining cyclophosphamide and double-stranded DNA preparations for eradication of Krebs-2 primary ascites in mice. Vavilov J Genet Breed. 2016; 20: 108-24[in Russian].

50

Penel N, Adenis A, Bocci G. Cyclophosphamide-based metronomic chemotherapy: after 10 years of experience, where do we stand and where are we going? Crit Rev Oncol Hematol. 2012; 82: 40-50.

51

Manrique SZ, Dominguez AL, Mirza N, Spencer CD, Bradley JM, Finke JH, et al. Definitive activation of endogenous antitumor immunity by repetitive cycles of cyclophosphamide with interspersed Tolllike receptor agonists. Oncotarget. 2016; 7: 42919-42.

52

Wu J, Waxman DJ. Metronomic cyclophosphamide schedule-dependence of innate immune cell recruitment and tumor regression in an implanted glioma model. Cancer Lett. 2014; 353: 272-80.

53

Doloff JC, Chen CS, Waxman DJ. Anti-tumor innate immunity activated by intermittent metronomic cyclophosphamide treatment of 9L brain tumor xenografts is preserved by anti-angiogenic drugs that spare VEGF receptor 2. Mol Cancer. 2014; 13: 1-13.

54

Ferrer-Font L, Arias-Ramos N, Lope-Piedrafita S, Julià-Sapé M, Pumarola M, Arús C, et al. Metronomic treatment in immunocompetent preclinical GL261 glioblastoma: effects of cyclophosphamide and temozolomide. NMR Biomed. 2017; 30: 1-12.

55

Ibe S, Qin Z, Schüler T, Preiss S, Blankenstein T. Tumor rejection by disturbing tumor stroma cell interactions. J Exp Med. 2001; 194: 1549-59.

56

Moskalenko M, Pan M, Fu Y, de Moll EH, Hashimoto D, Mortha A, et al. Requirement for innate immunity and CD90 NK1.1 lymphocytes to treat established melanoma with chemo-immunotherapy. Cancer Immunol Res. 2015; 3: 296-304.

57

Radojcic V, Bezak KB, Skarica M, Pletneva MA, Yoshimura K, Schulick RD, et al. Cyclophosphamide resets dendritic cell homeostasis and enhances antitumor immunity through effects that extend beyond regulatory T cell elimination. Cancer Immunol Immunother. 2010; 59: 137-48.

58

Salem ML, El-Naggar SA, Cole DJ. Cyclophosphamide induces bone marrow to yield higher numbers of precursor dendritic cells in vitro capable of functional antigen presentation to T cells in vivo. Cell Immunol. 2010; 261: 134-43.

59

Salem ML, Diaz-Montero CM, Al-Khami AA, El-Naggar SA, Naga O, Montero AJ, et al. Recovery from cyclophosphamide-induced lymphopenia results in expansion of immature dendritic cells which can mediate enhanced prime-boost vaccination antitumor responses in vivo when stimulated with the TLR3 agonist poly(I:C). J Immunol. 2009; 182: 2030-40.

60

Šefc L, Pšenák O, Sýkora V, Šulc K, Nečas E. Response of hematopoiesis to cyclophosphamide follows highly specific patterns in bone marrow and spleen. J Hematother Stem Cell Res. 2003; 12: 47-61.

61

Karman J, Ling C, Sandor M, Fabry Z. Dendritic cells in the initiation of immune responses against central nervous system-derived antigens. Immunol Lett. 2004; 92: 107-15.

62

Bauer M, Goldstein M, Heylmann D, Kaina B. Human monocytes undergo excessive apoptosis following temozolomide activating the ATM/ATR pathway while dendritic cells and macrophages are resistant. PLoS One. 2012; 7: 39956.

63

Won W-J, Deshane JS, Leavenworth JW, Oliva CR, Griguer CE. Metabolic and functional reprogramming of myeloid-derived suppressor cells and their therapeutic control in glioblastoma. Cell Stress. 2019; 3: 47-65.

64

Liu L, Taverna P, Whitacre CM, Chatterjee S, Gerson SL. Pharmacologic disruption of base excision repair sensitizes mismatch repair-deficient and -proficient colon cancer cells to methylating agents. Clin Cancer Res. 1999; 5: 2908-17.

Cancer Biology & Medicine
Pages 910-930
Cite this article:
Dolgova EV, Andrushkevich OM, Kisaretova PE, et al. Efficacy of the new therapeutic approach in curing malignant neoplasms on the model of human glioblastoma. Cancer Biology & Medicine, 2021, 18(3): 910-930. https://doi.org/10.20892/j.issn.2095-3941.2020.0511

48

Views

0

Downloads

5

Crossref

6

Web of Science

6

Scopus

Altmetrics

Received: 24 September 2020
Accepted: 08 February 2021
Published: 01 August 2021
©2021 Cancer Biology & Medicine.

Creative Commons Attribution-NonCommercial 4.0 International License

Return