Abstract

It is well established that the role of the tumor microenvironment (TME) in cancer progression and therapeutic resistance is crucial, but many of the underlying mechanisms are still being elucidated. Even with better understanding of molecular oncology and identification of genomic drivers of these processes, there has been a relative lag in identifying and appreciating the cellular drivers of both invasion and resistance. Intercellular communication is a vital process that unifies and synchronizes the diverse components of the tumoral infrastructure. Elucidation of the role of extracellular vesicles (EVs) over the past decade has cast a brighter light on this field. And yet even with this advance, in addition to diffusible soluble factor-mediated paracrine and endocrine cell communication as well as EVs, additional niches of intratumoral communication are filled by other modes of intercellular transfer. Tunneling nanotubes (TNTs), tumor microtubes (TMs), and other similar intercellular channels are long filamentous actin-based cellular conduits (in most epithelial cancer cell types, ~15-500 µm in length; 50–1000+ nm in width). They extend and form direct connections between distant cells, serving as conduits for direct intercellular transfer of cell cargo, such as mitochondria, exosomes, and microRNAs; however, many of their functional roles in mediating tumor growth remain unknown. These conduits literally create a physical bridge to create a syncytial network of dispersed cells amidst the intercellular stroma-rich matrix. Emerging evidence suggests that they provide a cellular mechanism for induction and emergence of drug resistance and contribute to increased invasive and metastatic potential. They have been imaged in vitro and also in vivo and ex vivo in tumors from human patients as well as animal models, thus not only proving their existence in the TME, but opening further speculation about their exact role in the dynamic niche of tumor ecosystems. TNT cellular networks are upregulated between cancer and stromal cells under hypoxic and other conditions of physiologic and metabolic stress. Furthermore, they can connect malignant cells to benign cells, including vascular endothelial cells. The field of investigation of TNT-mediated tumor-stromal, and tumor-tumor, cell-cell communication is gaining momentum. The mixture of conditions in the microenvironment exemplified by hypoxia-induced ovarian cancer TNTs playing a crucial role in tumor growth, as just one example, is a potential avenue of investigation that will uncover their role in relation to other known factors, including EVs. If the role of cancer heterocellular signaling via TNTs in the TME is proven to be crucial, then disrupting formation and maintenance of TNTs represents a novel therapeutic approach for ovarian and other similarly invasive peritoneal cancers.

Highlights

  • The tumor microenvironment (TME) is lush with a complex array of interacting parts

  • A recent comprehensive article by Worzfeld et al published in this journal discussed in detail the extreme molecular and cellular diversity of the ovarian cancer microenvironment [1]; this diversity presents a complex set of problems when it comes to translational application of knowledge of the ovarian TME and tumoral reaction in the face of biological insults stimulated by chemotherapeutic drugs and surgical resections

  • tunneling nanotubes (TNTs) have been imaged in intact tumors using confocal microscopy, demonstrating their potential in vivo relevance [23, 35, 47]. Investigators in this burgeoning field have proposed that TNTs/tumor microtubes (TMs) provide a physical and previously missing link in the chain of active players in cell communication in the dynamic and rapidly evolving TME, in ovarian as well as many other forms of invasive malignancy

Read more

Summary

Introduction

The tumor microenvironment (TME) is lush with a complex array of interacting parts. These parts include a multitude of cell types that evolve during the course of carcinogenesis and cancer cell progression. Let us examine ovarian and other peritoneal malignancies as paradigms for understanding the niche of TNTs in the TME, and their potential role in instigating cancer cell invasion, metastasis, and drug resistance.

Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call