Cancer stem cells and evolving novel therapies: a paradigm shift
Review Article

Cancer stem cells and evolving novel therapies: a paradigm shift

Sangeetha Vasudevaraj Naveen, Kumar Kalaivani

GLR Laboratories Private Limited, Mathur, Chennai, India

Contributions: (I) Conception and design: SV Naveen; (II) Administrative support: None; (III) Provision of study material: SV Naveen; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Dr. Sangeetha Vasudevaraj Naveen. GLR Laboratories Private Limited, 444 Gokulam Street, Mathur, Chennai 600068, India. Email: sangeevv@gmail.com.

Abstract: Accumulating evidence of stem-like cells/cancer stem cells (CSCs) has been gaining attention of cancer researchers over the last decade. Though many tumors harbor CSCs in their dedicated niches, identifying and exterminating those cells has proved to be difficult, due to their heterogenous nature, as the CSC phenotype vary substantially and may undergo reversible phenotypic changes. As a tumor propagation initiator, CSCs are considered as an exciting novel therapy for a better therapeutic outcome. This review discusses the major advances in the development of CSC-based therapies of most common cancers which includes lung, cervix and liver cancers.

Keywords: Cancer stem cells (CSCs); lung cancer; cervix cancer; liver cancer; nanoparticles (NPs); novel therapies


Received: 05 December 2017; Accepted: 26 December 2017; Published: 23 January 2018.

doi: 10.21037/sci.2018.01.03


Introduction

Cancer is an umbrella term covering a plethora of conditions characterized by unscheduled and uncontrolled cellular proliferation. As the average life expectancy in many countries steadily rises, so do cancer-related deaths, thus making cancer as most common causes of death in 21st century. Despite several debates on the causes of disease, the history of medicine teaches us the need for understanding the scientific basis before the development of successful therapy. The paradigm shift in cancer treatment, with surgery, radiotherapy and conventional cytotoxic chemotherapy for the past 20 years has made only a modest overall impact on mortality. Though, childhood cancers, testicular cancer and lymphoma can be cured, and the survival rates of breast and colorectal cancer have been improved through adjuvant drug treatment, majority of human cancers are difficult to treat, especially in their advanced, metastatic forms (1). There is thus a pressing need for novel and effective forms of systemic therapy.

As a result of the advanced research, evidences suggest that the relapse of the disease depends on the small subset of cells within the tumor, termed as cancer progenitor cells/cancer stem cells (CSCs) or cancer initiating cells (Figure 1).

Figure 1 An overview of emerging therapeutic concepts in CSC biology. CSC, cancer stem cell; ABC, ATP-binding cassette.

Though intensively researched, much remains to be elucidated about their function in initiation and progression to metastatic disease states and resistance to conventional therapies. The self-renewal ability, aberrant multi-lineage differentiation potential and the ‘stemness’ property of CSCs is said to contribute, to the formation of the heterogeneous cellular population into major cell types observed in the corresponding tumor, which is resistant to ROS or DNA damage (1,2). Furthermore, occurrence and recurrence of highly aggressive cancer subtypes may transpire, owing to the accumulation of different genetic and/or epigenetic alterations in cancer progenitor cells during cancer progression, which in turn has inspired the design of innovative treatment strategies for most common types of cancers, including leukemia (3,4), breast cancer (5), colorectal cancer (6,7) and brain cancer (1,8) which now aims at exterminating CSCs rather than shrinking tumor bulk.

Since, recent evidence suggests that CSCs are resistant to radio and chemotherapy (9), the CSC model became the foundation for new preventive and therapeutic strategies in cancer. Intensified understanding of CSC-derived heterogeneity, the asymmetric division of CSCs which enables the cells to generate differentiated progeny, will provide additional insights to efficiently eliminate CSCs through targeting CSC-marker, CSC-specific cellular signaling pathways, and CSC-microenvironment (10,11). The aim of this review is to focus on major advances in the development of CSC-based therapies of most common cancers which includes lung, cervix and liver cancers.


Lung cancer: the leading killer

Lung cancer, classified as non-small cell lung cancer (NSCLC) and small-cell lung cancer (SCLC), is the leading cause of cancer-related death with highest morbidity and mortality in the United States (12,13). Despite novel molecular therapies, the prognosis of NSCLC, which accounts for 85% of cases, has low treatment response rates and poor overall prognosis (1% estimated survival rate). If diagnosed early, NSCLC patients may benefit from surgery and result in a cure, but, SCLC are almost never diagnosed early, even after surgery rarely result in a cure.

Standard chemotherapy and radiotherapy, has reached a plateau of effectiveness in improving survival. Therapies targeting new blood vessels, treatments interfering with chemical signal required for cancer cell growth, treatments targeting the receptors like epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) are being tried (14,15). Cascone et al., has reported that the combined use of EGFR-tyrosine kinase inhibitor erlotinib and the humanized VEGF receptor monoclonal antibody bevacizumab in advanced, chemotherapy-refractory NSCLC has shown promising results (16).

Arrival of targeted therapies harboring BRAF, MET, HER2 and RET alterations, understanding early epigenetic targets and advances in understanding the role of DNA methylation and histone modifiers have established new standards of care for defined molecular subsets of NSCLC (17). In lung cancer, CSC populations have been identified and enriched in multiple phenotypic sub-population, such as drug-resistant side population (SP) (18) CD133pos cells (19,20) and ALDH high cells (21,22) and exhibit plasticity. An independent study demonstrated an increase in the fraction of CD133 positive lung cancer cells present after treatment of mice carrying human lung cancer xenografts with cisplatin (23). CSC possess several mechanisms to overcome irreversible damage by cytostatic drugs, thereby has been claimed as chemo resistant. Strong evidence for the presence of drug-resistant and highly regenerative tumor cells in NSCLC has been reported (24,25). However, Chandrakesan et al., has reported that the combination therapy of siDCLKL1 (doublecortin like kinase 1) with cisplatin can overcome cisplatin induced drug resistance thus making DCLK1 targeted therapy an attractive tool for combating NSCLC (26). Also, novel imaging techniques are being developed to identify and track these stem cells in their natural niche may provide better understanding on the significance of CSCs in drug resistance, tumor regeneration and metastasis.

CSC drugs targeting stem-like traits of cancer cells could be effective in controlling refractory EML4-ALK+ NSCLC (25), along with the signaling pathways which are critically involved in CSC regulation. Currently, several early phase studies analyzing the efficacy of Wnt, Notch, MAPK and Hedgehog pathways as well as the PI3K/AKT/mTOR signal transduction axis are in progress.

Though, several fundamental results on lung stem cells were derived from murine models, whether the findings can fit human conditions, are still a debate. Meanwhile, the therapeutic management of patients with advanced NSCLC is an ongoing challenge and several novel agents targeting cancer are under investigation.


Cervical cancer and CSCs

Among the most commonly diagnosed cancers in women, cervical cancer accounts for 7.9% and occupies the second and third position as the leading cause of cancer-related death worldwide. Whilst the treatment strategies include surgery and chemotherapy, it doesn’t provide a permanent cure. Whereas, radical surgery for advanced stage affects the child bearing ability of the patient, along with risk of recurrence (27). Currently, hysterectomy and radiation therapy are used to treat and cure cervical cancer at early stage (28). The gene expression profiling of cervical cancer cells has shown aberrant methylations of the CpG island within the promoters of several tumor-suppressor genes including p53, which is normally involved in the positive regulation of apoptosis and negative regulation of cell growth and migration (29). Studies have shown that approximately 90% of CIN3 (abnormality of squamous cells lined in ectocervix) and cervical cancer arise within or very near the SC junction, where the malignant lesions are initiated by an interaction between human papilloma virus (HPV) infection and the genetic and epigenetic alteration of healthy stem cell (30). These CSCs bypass drug cytotoxicity, by using Adenosine triphosphate (ATP)-dependent protein family’s efflux pumps (31), thereby marking ATP-binding cassette sub-family G member 2 (ABCG2) as a potential target. Studies has also revealed several other markers along with ABCG2, which includes ALDH1A1, CD133, CK-17, p63+, AII+, CD49F, OCT4, osteopontin (OPN), SOX-2, NANOG, CD44, C-KIT (32). However, the detailed mechanisms responsible for higher expression of ALDH1A1 in cervical cancer stem cells (CCSCs) are largely unknown. Liu et al. found that the cervical cancer cells, which were resistant to radiotherapy, exhibited a higher percentage of surface markers CD44+ and CD24+ (33). Studies also showed a critical role of AP-1, a key regulator for expression of HPV oncogenes, in mediating chemo- and/or radioresistance (34,35). Tyagi et al., have targeted AP-1 with herbal derivative curcumin to induce radio-sensitization and downregulated c-Fos, c-Jun and upregulated Fra-1 for effective cancer treatment (36).

Though CSC-targeted therapies have been intensively studied, specific CCSC targeted therapies are very limited. Likewise, dual-targeting strategies, for example VS-5584, as a potent and selective dual inhibitor of mTORC1/2 and class I PI 3-kinases (PI3K), which specifically targets human CSCs have been reported (37), however, no studies have yet reported on the use of dual-targeting to treat CCSCs.


Liver cancer and CSCs

Liver cancer, the fifth most prevalent cancer and third leading cause of cancer related death is associated with hepatitis C virus (HCV) infection along with HBV. Among primary liver cancers, hepatocellular carcinoma (HCC) represents the major histological subtype, accounting for 70–85% of cases of primary liver cancer (38). Intrahepatic cholangiocarcinoma (ICC) is the second most frequent type of liver cancer, and its incidence has been increasing. Reports say that Western’s population develops HCC in cirrhotic liver while noncirrhotic liver cancer develops in 50% of Asian countries. Metabolic syndrome is said to be an additional risk factor for HCC development (39).

Among others, therapeutic options and prognosis of the patients mainly depends on the stage on the presentation, based on several prognostic factors, which includes tumour size, number of the lesions, liver remnant function. Several markers including lens culinaris agglutinin-reactive AFP (AFP-L3), des-carboxyprothrombin (DCP), glypican-3 (GPC-3), OPN, and several other biomarkers (such as squamous cell carcinoma antigen immunoglobulin M complexes, alpha-1-fucosidase (AFU), chromogranin A (CgA), human hepatocyte growth factor, and insulin-like growth factor (IGF) have been proposed for the early detection of HCC (40). Though none of them is optimal, when used together, their sensitivity in detecting HCC may increase.

Earlier stages of HCC are treated with resection or liver transplantation; however, tumor recurrence rate is still high up to 70%. Takayama et al. has proposed that adoptive immunotherapy, anti-CD3 and IL-2 stimulated autologous T lymphocytes infused in HCC patients may significantly improve postsurgical recurrence-free survival (41). Radiofrequency ablation (RFA), microwave ablation (MWA) or transarterial chemoembolism (TACE) were used to treat the intermediate stages. For advanced patients with large non-resectable lesions, Sorafenib is the standard of care. Sunitinib malate, an oral multikinase inhibitor targets several tyrosine kinases receptors, such as VEGF-1/2 and PDGFR-a/b, and is implicated in HCC proliferation and angiogenesis. However, sunitinib seems to have more side effects for its toxicity in HCC. Along with multikinase inhibitors, MET inhibitors, Antiangiogenic agents and mTOR inhibitors have been tested in HCC with marginal efficacy to date (42).

Recent insights into the molecular pathogenesis of HCC have identified several aberrant signaling pathways that have served as targets for novel therapeutic agents. Several pathways are now implicated in hepatocarcinogenesis and agents that target these pathways continue to be developed. For example, RAS/RAF/MAPK pathway is typically activated in HCC as a result of increased signaling induced from upstream growth factors and due to inactivation of tumor suppressor genes (43). Though Sorafenib, significantly inhibit RAS/RAF/MAPK pathway, the heterogenicity of these cancers may warrant a combination therapy for advanced stage HCC. Several reagents are being tested targeting novel signaling cascades such as Wnt-β-catenin and Notch.

Lately, the focus of treatment has shifted to explore the possibilities of inhibiting liver cancer stem cells (LCSCs). Tumor markers have been a mainstay of identifying cancer cells in all tissues. Lui et al., has reported that LCSCs can be recognized by multiple cell surface antigens including CD133, CD90, CD44, CD24, and the epithelial cell adhesion molecule (EpCAM) (44).

Doublecortin like kinase protein 1 (Dclk1) is considered to be an important target for the treatment of tumors of the liver, pancreas, and colon. Studies were able to show that these Dclk1+ cells possess features of cancer stem (initiating) cells (45-47). In addition to OCT4, c-myc, NANOG and Sox-9, zinc finger transcription factors have been reported to regulate liver CSCs features. ZIC2, has been demonstrated to be highly expressed in liver CSCs which regulates and maintain liver CSC self-renewal by recruiting NURF complex to trigger OCT4 activation (48). miRNAs and lncRNAs (49) play an important role in regulating the properties of liver CSCs and therefore could be therapeutic targets. Epigenetic alterations, including DNA methylation, histone modifications, polycomb repressive complex (PRC), and chromatin remodeling complex function, are mechanisms that contribute directly to carcinogenesis and CSC regulation. Inhibiting histone deacetylase SIRT1, EZH2 can be a promising therapeutic approach to eradicate liver CSCs.


Conclusions

Accumulating studies, over the decade, supports the existence of CSCs in many cancer types, thus modifying our perception of cancer cure. Though complicated, new insights in to stem cell biology helps us to understand the features and behaviors of CSCs. Development of new technologies, opens up many avenues for analyzing CSCs in their intact environment. For example, CSCs targeting nanoparticles (NPs) is the topic of recent investigations, where the NPs is either with a ligand/cytotoxic anticancer drug/a chemosensitizer (such as ABC transporter inhibitor) or an imaging agent which facilitates tumor diagnostics. However, the identification of strategies that exploit the unique characteristics of CSCs requires further study and the cooperation of multidisciplinary areas.


Acknowledgements

The authors would like to thank Dr. T.S. Kumaravel and Dr. S.S. Murugan of GLR Laboratories Private Limited, for provision of necessary facilities in the preparation of this manuscript.


Footnote

Conflicts of Interest: The authors have no conflicts of interest to declare.


References

  1. Batlle E, Clevers H. Cancer stem cells revisited. Nat Med 2017;23:1124-34. [Crossref] [PubMed]
  2. Mimeault M, Hauke R, Batra SK. Stem cells: a revolution in therapeutics-recent advances in stem cell biology and their therapeutic applications in regenerative medicine and cancer therapies. Clin Pharmacol Ther 2007;82:252-64. [Crossref] [PubMed]
  3. Uckun FM, Sather H, Reaman G, et al. Leukemic cell growth in SCID mice as a predictor of relapse in high-risk B-lineage acute lymphoblastic leukemia. Blood 1995;85:873-8. [PubMed]
  4. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med 1997;3:730-7. [Crossref] [PubMed]
  5. Al-Hajj M, Wicha MS, Benito-Hernandez A, et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003;100:3983-8. [Crossref] [PubMed]
  6. O'Brien CA, Pollett A, Gallinger S, et al. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007;445:106-10. [Crossref] [PubMed]
  7. Ricci-Vitiani L, Lombardi DG, Pilozzi E, et al. Identification and expansion of human colon-cancer-initiating cells. Nature 2007;445:111-5. [Crossref] [PubMed]
  8. Singh SK, Hawkins C, Clarke ID, et al. Identification of human brain tumour initiating cells. Nature 2004;432:396-401. [Crossref] [PubMed]
  9. Ricardo S, Vieira AF, Gerhard R, et al. Breast cancer stem cell markers CD44, CD24 and ALDH1: expression distribution within intrinsic molecular subtype. J Clin Pathol 2011;64:937-46. [Crossref] [PubMed]
  10. Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell 2014;14:275-91. [Crossref] [PubMed]
  11. Matsuda S, Yan T, Mizutani A, et al. Cancer stem cells maintain a hierarchy of differentiation by creating their niche. Int J Cancer 2014;135:27-36. [Crossref] [PubMed]
  12. Jemal A, Siegel R, Xu J, et al. Cancer statistics, 2010. CA Cancer J Clin 2010;60:277-300. [Crossref] [PubMed]
  13. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359-86. [Crossref] [PubMed]
  14. Olaussen KA, Dunant A, Fouret P, et al. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med 2006;355:983-91. [Crossref] [PubMed]
  15. Ceppi P, Volante M, Novello S, et al. ERCC1 and RRM1 gene expressions but not EGFR are predictive of shorter survival in advanced non-small-cell lung cancer treated with cisplatin and gemcitabine. Ann Oncol 2006;17:1818-25. [Crossref] [PubMed]
  16. Cascone T, Gridelli C, Ciardiello F. Combined targeted therapies in non-small cell lung cancer: a winner strategy? Curr Opin Oncol 2007;19:98-102. [Crossref] [PubMed]
  17. Lau KW, Seng C, Lim TKH, et al. Expanded molecular interrogation for potential actionable targets in non-squamous non-small cell lung cancer. Ann Transl Med 2017;5:372. [Crossref] [PubMed]
  18. Ho MM, Ng AV, Lam S, et al. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Res 2007;67:4827-33. [Crossref] [PubMed]
  19. Eramo A, Lotti F, Sette G, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death Differ 2008;15:504-14. [Crossref] [PubMed]
  20. Levina V, Marrangoni AM, DeMarco R, et al. Drug-selected human lung cancer stem cells: cytokine network, tumorigenic and metastatic properties. PLoS One 2008;3:e3077. [Crossref] [PubMed]
  21. Deng S, Yang X, Lassus H, et al. Distinct expression levels and patterns of stem cell marker, aldehyde dehydrogenase isoform 1 (ALDH1), in human epithelial cancers. PLoS One 2010;5:e10277. [Crossref] [PubMed]
  22. Sullivan JP, Spinola M, Dodge M, et al. Aldehyde dehydrogenase activity selects for lung adenocarcinoma stem cells dependent on notch signaling. Cancer Res 2010;70:9937-48. [Crossref] [PubMed]
  23. Bertolini G, Roz L, Perego P, et al. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc Natl Acad Sci U S A 2009;106:16281-6. [Crossref] [PubMed]
  24. Garcia Campelo MR, Alonso Curbera G, Aparicio Gallego G, et al. Stem cell and lung cancer development: blaming the Wnt, Hh and Notch signalling pathway. Clin Transl Oncol 2011;13:77-83. [Crossref] [PubMed]
  25. Gottschling S, Schnabel PA, Herth FJ, et al. Are we missing the target? Cancer stem cells and drug resistance in non-small cell lung cancer. Cancer Genomics Proteomics 2012;9:275-86. [PubMed]
  26. Chandrakesan P, Qu D, May R, et al. DCLK1 a novel therapeutic target in non-small cell lung cancer. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017. Philadelphia: AACR 2017;77.
  27. Chhabra R. Cervical cancer stem cells: opportunities and challenges. J Cancer Res Clin Oncol 2015;141:1889-97. [Crossref] [PubMed]
  28. Shrivastava SK, Mahantshetty U, Narayan K. Principles of radiation therapy in low-resource and well-developed settings, with particular reference to cervical cancer. Int J Gynaecol Obstet 2015;131 Suppl 2:S153-8. [Crossref] [PubMed]
  29. Vande Pol SB, Klingelhutz AJ. Papillomavirus E6 oncoproteins. Virology 2013;445:115-37. [Crossref] [PubMed]
  30. Lopez J, Ruiz G, Organista-Nava J, et al. Human papillomavirus infections and cancer stem cells of tumors from the uterine cervix. Open Virol J 2012;6:232-40. [Crossref] [PubMed]
  31. Cui H, Zhang AJ, Chen M, et al. ABC Transporter Inhibitors in Reversing Multidrug Resistance to Chemotherapy. Curr Drug Targets 2015;16:1356-71. [Crossref] [PubMed]
  32. Huang R, Rofstad EK. Cancer stem cells (CSCs), cervical CSCs and targeted therapies. Oncotarget 2017;8:35351-67. [PubMed]
  33. Liu H, Wang YJ, Bian L, et al. CD44+/CD24+ cervical cancer cells resist radiotherapy and exhibit properties of cancer stem cells. Eur Rev Med Pharmacol Sci 2016;20:1745-54. [PubMed]
  34. Dhandapani KM, Mahesh VB, Brann DW. Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFkappaB transcription factors. J Neurochem 2007;102:522-38. [Crossref] [PubMed]
  35. Kajanne R, Miettinen P, Tenhunen M, et al. Transcription factor AP-1 promotes growth and radioresistance in prostate cancer cells. Int J Oncol 2009;35:1175-82. [PubMed]
  36. Tyagi A, Vishnoi K, Kaur H, et al. Cervical cancer stem cells manifest radioresistance: Association with upregulated AP-1 activity. Sci Rep 2017;7:4781. [Crossref] [PubMed]
  37. Kolev VN, Wright QG, Vidal CM, et al. PI3K/mTOR dual inhibitor VS-5584 preferentially targets cancer stem cells. Cancer Res 2015;75:446-55. [Crossref] [PubMed]
  38. Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA Cancer J Clin 2011;61:69-90. [Crossref] [PubMed]
  39. Simard EP, Ward EM, Siegel R, et al. Cancers with increasing incidence trends in the United States: 1999 through 2008. CA Cancer J Clin 2012;62:118-28. [Crossref] [PubMed]
  40. Yan J, Yang X, Li L, et al. Low expression levels of insulin-like growth factor binding protein-3 are correlated with poor prognosis for patients with hepatocellular carcinoma. Oncol Lett 2017;13:3395-402. [Crossref] [PubMed]
  41. Takayama T, Sekine T, Makuuchi M, et al. Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet 2000;356:802-7. [Crossref] [PubMed]
  42. Bertino G, Demma S, Ardiri A, et al. Hepatocellular carcinoma: novel molecular targets in carcinogenesis for future therapies. Biomed Res Int 2014;2014:203693. [Crossref] [PubMed]
  43. Galuppo R, Ramaiah D, Ponte OM, et al. Molecular therapies in hepatocellular carcinoma: what can we target? Dig Dis Sci 2014;59:1688-97. [Crossref] [PubMed]
  44. Lui SK, Vilchez V, Gedaly R. Liver Cancer Stem Cells: A New Paradigm for Hepatocellular Carcinoma Treatment. J Stem Cell Res Ther 2015;5:283.
  45. Westphalen CB, Quante M, Wang TC. Functional implication of Dclk1 and Dclk1-expressing cells in cancer. Small GTPases 2017;8:164-71. [Crossref] [PubMed]
  46. Nguyen CB, Kotturi H, Waris G, et al. (Z)-3,5,4'-Trimethoxystilbene Limits Hepatitis C and Cancer Pathophysiology by Blocking Microtubule Dynamics and Cell-Cycle Progression. Cancer Res 2016;76:4887-96. [Crossref] [PubMed]
  47. Chandrakesan P, Weygant N, Qu D, et al. Knocking down the expression of DCLK1 reduces mammary tumor formation, tumor cell self-renewal and metastasis: DCLK1 a novel therapeutic target in breast cancer. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research. Philadelphia: AACR, 2016;76.
  48. Zhu P, Wang Y, He L, et al. ZIC2-dependent OCT4 activation drives self-renewal of human liver cancer stem cells. J Clin Invest 2015;125:3795-808. [Crossref] [PubMed]
  49. Zhu P, Wang Y, Wu J, et al. LncBRM initiates YAP1 signalling activation to drive self-renewal of liver cancer stem cells. Nat Commun 2016;7:13608. [Crossref] [PubMed]
doi: 10.21037/sci.2018.01.03
Cite this article as: Naveen SV, Kalaivani K. Cancer stem cells and evolving novel therapies: a paradigm shift. Stem Cell Investig 2018;5:4.

Download Citation