Cancer Associated Fibroblasts Review

Advertisement

Introduction


Cancer-associated fibroblasts (CAFs) are a predominant component of the tumor microenvironment (TME) and play a critical role in tumor progression, metastasis, and therapy resistance. Historically considered as passive structural elements, CAFs are now recognized as dynamic and heterogeneous cells that actively communicate with cancer cells and other stromal components. Their complex interactions influence tumor growth, angiogenesis, immune evasion, and response to treatment. This review aims to provide a comprehensive overview of CAFs, including their origin, phenotypic characteristics, functional roles, heterogeneity, and potential therapeutic strategies targeting these cells.

Origins and Activation of Cancer-associated Fibroblasts



Sources of CAFs


CAFs can originate from multiple cell types within the tissue microenvironment, including:

  • Resident fibroblasts: Quiescent fibroblasts in normal tissue can become activated upon tumor-associated stimuli.

  • Epithelial-to-mesenchymal transition (EMT): Epithelial cells can undergo EMT, acquiring fibroblast-like features.

  • Endothelial-to-mesenchymal transition (EndMT): Endothelial cells may transition into CAF-like cells.

  • Bone marrow-derived mesenchymal stem cells (MSCs): Circulating MSCs can migrate into tumors and differentiate into CAFs.

  • Pericytes and other stromal components: These cells may also contribute to the CAF pool under tumor-promoting conditions.



Mechanisms of Activation


The activation of fibroblasts into CAFs is driven by various tumor-derived factors, including:
- Transforming growth factor-beta (TGF-β)
- Platelet-derived growth factor (PDGF)
- Fibroblast growth factor (FGF)
- Interleukins such as IL-6 and IL-8
- Extracellular matrix (ECM) stiffness and remodeling signals

These factors induce the expression of activation markers, such as alpha-smooth muscle actin (α-SMA), fibroblast activation protein (FAP), and platelet-derived growth factor receptor (PDGFR), transforming normal fibroblasts into an activated, tumor-promoting phenotype.

Phenotypic Characteristics of CAFs


CAFs are characterized by a diverse set of markers and functional properties that distinguish them from normal fibroblasts. However, their heterogeneity poses challenges in defining a universal marker.

Common Markers of CAFs


Some of the frequently used markers include:

  • Alpha-smooth muscle actin (α-SMA)

  • Fibroblast activation protein (FAP)

  • Platelet-derived growth factor receptors (PDGFR-α and PDGFR-β)

  • Vimentin

  • Tenascin-C

  • Fibroblast-specific protein 1 (FSP1/S100A4)



It is important to note that no single marker is exclusively specific to CAFs, and their expression can overlap with other stromal or mesenchymal cell types.

Heterogeneity of CAFs


Emerging evidence indicates that CAFs are a heterogeneous population with distinct phenotypic and functional subsets. These subsets can be classified based on their gene expression profiles, localization within tumors, and roles in tumor biology. Major subtypes include:

  1. Myofibroblastic CAFs (myCAFs): Characterized by high α-SMA expression, these cells are primarily involved in ECM remodeling and stiffening.

  2. Inflammatory CAFs (iCAFs): Secrete cytokines and chemokines like IL-6, contributing to inflammation and immunomodulation.

  3. Antigen-presenting CAFs (apCAFs): Express MHC class II molecules and may participate in immune regulation.



Understanding this heterogeneity is vital for designing targeted therapies and predicting tumor behavior.

Functional Roles of CAFs in Tumor Progression



ECM Remodeling and Tumor Stiffness


CAFs produce and modify ECM components, including collagen, fibronectin, and hyaluronan, leading to increased matrix stiffness. This remodeling facilitates:
- Enhanced tumor cell invasion
- Creation of tracks for migration
- Elevated interstitial pressures that hinder drug delivery

Promotion of Tumor Cell Growth and Survival


CAFs secrete growth factors such as:
- Hepatocyte growth factor (HGF)
- Insulin-like growth factors (IGFs)
- FGF
- TGF-β

These factors support proliferation, survival, and stemness of cancer cells, fostering tumor expansion.

Angiogenesis


CAFs contribute to neovascularization by releasing angiogenic factors like VEGF, PDGF, and FGF, which stimulate endothelial cell proliferation and new blood vessel formation, ensuring adequate nutrient and oxygen supply to the tumor.

Immune Modulation and Evasion


CAFs modulate the immune landscape within tumors by:
- Secreting immunosuppressive cytokines such as IL-6, TGF-β, and CXCL12
- Recruiting immunosuppressive cells like regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs)
- Downregulating cytotoxic immune responses, thereby facilitating immune evasion

Metastasis Facilitation


CAFs promote metastasis via:
- Inducing epithelial-mesenchymal transition (EMT) in cancer cells
- Creating a pre-metastatic niche through ECM deposition and secretion of soluble factors
- Enhancing tumor cell motility and invasion

Heterogeneity and Plasticity of CAFs


Understanding CAF heterogeneity is crucial because different subsets may have opposing roles—some may promote tumor progression, while others might exert tumor-restraining effects. The plasticity of CAFs allows them to switch phenotypes in response to environmental cues, complicating therapeutic targeting.

Recent single-cell sequencing studies have unveiled diverse CAF subsets across tumor types, emphasizing the importance of context-specific approaches to targeting these cells.

Therapeutic Strategies Targeting CAFs



Direct Targeting of CAFs


Strategies include:
- FAP inhibitors or antibodies
- PDGFR blockade
- Depleting CAF populations using inducible genetic models

However, indiscriminate depletion of CAFs may lead to unintended consequences, such as increased tumor aggressiveness, highlighting the need for precise targeting.

Modulating CAF Phenotypes


Instead of depletion, reprogramming CAFs into a quiescent or tumor-restraining state is an emerging approach. Agents such as all-trans retinoic acid (ATRA) and vitamin D analogs have shown promise in reverting CAF activation.

Targeting CAF-Mediated ECM Remodeling


Inhibitors of matrix metalloproteinases (MMPs) and lysyl oxidase (LOX) can reduce ECM stiffness, improving drug delivery and reducing invasion.

Interrupting CAF-Tumor Cell Communication


Blocking key signaling pathways, such as TGF-β or HGF/c-Met, can disrupt pro-tumorigenic interactions.

Combination Therapies


Combining CAF-targeted therapies with chemotherapy, immunotherapy, or anti-angiogenic agents may enhance overall treatment efficacy.

Challenges and Future Perspectives


Despite advances, several challenges remain:
- Heterogeneity of CAF populations complicates targeting
- Lack of specific markers for selective CAF subset targeting
- Potential for adverse effects when depleting or modulating CAFs
- Limited understanding of CAF plasticity and reprogramming mechanisms

Future research should focus on:
- Developing precise molecular markers for CAF subsets
- Understanding the context-dependent roles of CAFs
- Designing combination therapies that modulate the tumor stroma without promoting tumor progression
- Leveraging advanced technologies like single-cell sequencing and spatial transcriptomics

Conclusion


Cancer-associated fibroblasts are integral components of the tumor microenvironment, exerting multifaceted effects on tumor progression, immune evasion, and therapy resistance. Their heterogeneity and plasticity present both challenges and opportunities for therapeutic intervention. A nuanced understanding of CAF biology, coupled with innovative targeting strategies, holds promise for improving cancer treatment outcomes. As research continues to unravel the complexities of CAFs, integrating stromal targeting with existing therapies may become a cornerstone of precision oncology in the future.

Frequently Asked Questions


What are cancer-associated fibroblasts (CAFs) and what role do they play in tumor progression?

Cancer-associated fibroblasts (CAFs) are a prominent component of the tumor microenvironment that support tumor growth, invasion, and metastasis by secreting growth factors, remodeling the extracellular matrix, and modulating immune responses.

What are the latest therapeutic strategies targeting CAFs in cancer treatment?

Recent approaches include targeting CAF-specific markers, disrupting their signaling pathways, reprogramming CAFs to a tumor-inhibitory phenotype, and combining CAF-targeted therapies with immunotherapy or chemotherapy to enhance treatment efficacy.

How do CAFs influence immune cell infiltration and immune evasion in tumors?

CAFs can create an immunosuppressive microenvironment by secreting cytokines and chemokines that inhibit immune cell infiltration, promote regulatory immune cells, and modulate immune checkpoints, thereby facilitating tumor immune evasion.

What are the challenges in identifying and targeting CAF heterogeneity within tumors?

The heterogeneity of CAF populations, with diverse origins and functions, complicates their identification and targeted therapy, as some subsets may have tumor-promoting roles while others could be tumor-suppressive, necessitating precise characterization.

What insights have recent reviews provided regarding the molecular mechanisms driving CAF activation?

Recent reviews highlight key signaling pathways such as TGF-β, PDGF, and Notch, as well as epigenetic modifications, that drive CAF activation and sustain their pro-tumorigenic functions, offering potential molecular targets for intervention.

How has the understanding of CAFs evolved in recent cancer research reviews?

Recent reviews have shifted from viewing CAFs as a uniform population to recognizing their heterogeneity and complex roles, emphasizing the importance of context-dependent functions and the potential for selective targeting to improve therapeutic outcomes.