Open access peer-reviewed chapter

Perspective Chapter: Role of Cancer-Associated Fibroblasts in Oncogenesis

Written By

Anyu Gu, Chikezie O. Madu and Yi Lu

Submitted: 27 August 2022 Reviewed: 31 October 2022 Published: 04 January 2023

DOI: 10.5772/intechopen.108832

From the Edited Volume

Tumor Microenvironment - New Insights

Edited by Ahmed Lasfar

Chapter metrics overview

77 Chapter Downloads

View Full Metrics

Abstract

The tumor microenvironment consists of multiple types of cells, including endothelial cells, pericytes, neutrophil macrophage mast cells, lymphatic cells, basement membrane extracellular matrix, as well as fibroblasts. Fibroblasts populations found in cancers, also known as cancer-associated fibroblasts, have been implicated in the initiation, progression, and metastasis of tumors. This chapter will focus on the roles of cancer-associated fibroblasts in the progression of cancer and the studies of use of cancer-associated fibroblasts as a therapeutic target for cancer intervention.

Keywords

  • tumor microenvironment
  • cancer-associated fibroblasts
  • fibroblasts
  • cancer intervention
  • cancer

1. Introduction

The tumor microenvironment (TME) is the environment in which tumor cells or cancer stem cells exist [1]. The TME consists of multiple types of cells, including endothelial cells, immune cells, and fibroblasts [1, 2, 3]. The TME also consists of components such as the extracellular matrix (ECM), soluble factors such as cytokines and growth factors, and physical properties such as pH and oxygen content [2]. The TME and the interactions between its components help to promote tumor growth and cancer progression (Figure 1) [3].

Figure 1.

Important interactions and mechanisms of the TME [1].

Fibroblasts are the most common type of cell in connective tissue, commonly defined as structural cells that specialize in depositing and remodeling the ECM [4]. Fibroblast populations found in primary and metastatic cancers, known as cancer-associated fibroblasts (CAFs), are implicated in tumor initiation, progression, and metastasis [5]. CAFs have wide varieties of cells-of-origin, heterogeneous phenotypes, and diverse functions, all of which are shared by other cells found in the TME [6]. This chapter will focus on CAFs and their potential use in cancer intervention.

Advertisement

2. Fibroblasts and cancer-associated fibroblasts

The precursors of CAFs are generally considered to be dormant tissue-resident fibroblasts and pancreatic and hepatic stellate cells, though different studies have also identified bone marrow-derived mesenchymal stem cells, endothelial cells, and adipocytes [5]. Fibroblasts play a prominent role in coordinating the wound repair response in skin; therefore, it is likely that key CAF traits correspond to the normal physiological role normal fibroblasts play [7]. Fibroblasts transform into CAFs through tumor-derived stimuli, including soluble factors secreted by the tumor, immune infiltrate, lysophosphatidic acid, fibroblast growth factor, interleukin-1 (IL-1), IL-6, and granulin [8]. Transforming growth factor β (TGFβ) and lysophosphatidic acid are well-established activating signals for fibroblasts, which promote the activity of SMAD transcription factors and serum response factors, respectively [7]. These fibroblast activating signals converge to drive expression of the fibroblast marker αSMA, as well as increase the activity of the contractile cytoskeleton [7]. Fibroblasts may become activated through Notch signaling when in direct contact with tumor cells [7, 8]. Other mechanisms that can activate normal fibroblasts to become CAFs are shown in Figure 2. In the TME, tumor cells secrete factors such as TGFβ, platelet-derived growth factor (PGDF), and fibroblast growth factor (FGF) to convert fibroblasts to CAFs [3]. A build-up of CAFs is often associated with poor prognosis in many cancer types [3].

Figure 2.

Mechanisms that activate normal fibroblasts to become CAFs. FGF, fibroblast growth factor; PDGF, platelet-derived growth factor; ROS, reactive oxygen species; RTK, receptor tyrosine kinase; TGFβ, transforming growth factor-β; TNF, tumor necrosis factor [7].

As shown in Figure 3 [10], TGFβ is a common factor in the conversion of many different cell types into fibroblasts and CAFs. There are many types of TGFβ. TGFβ-1 is one that is secreted by stromal and tumor cells and is the main factor in promoting the mobilization of residential fibroblasts and their activation into CAFs [10]. Through SMAD-dependent and SMAD-independent pathways, TGFβ-1 activates fibroblasts into CAFs, expressing alpha-smooth muscle actin, periostin, α-fibroblast activation protein, and fibroblast-specific protein-1 [10]. In addition, activated fibroblasts secrete TGFβ-1 [10], which could create a positive feedback loop, increasing fibroblast activation. TGFβ binds to the type 2 of TGFβ receptor (TGFBR2) on the surface of fibroblasts [11].

Figure 3.

Different origins of CAFs [9, 10].

2.1 CAFs in tumors

CAFs are a type of myofibroblast that enhance the malignancy and progression of cancer [12]. The presence of CAFs is identified in almost all solid tumors [13]. This suggests that CAFs are important to the formation of solid tumors. In an established tumor, the TME represents a changed part of the original normal tissue of the host [13]. Tumor cells mostly contribute to the change in their favor. [13]. The stromal transformation of TME is primarily dominated and maintained by CAFs [13]. The CAF component of the TME is the most critical in influencing most of the functions of the TME in real time [13]. CAFs alter the TME by directly interacting with cancer cells and regulatory paracrine signaling, control the immune response to neoplasia, deposit ECM components, stimulate angiogenesis, and provide a scaffold for tumor invasion and metastasis [14]. Additionally, CAFs can produce many growth factors and pro-inflammatory cytokines to promote angiogenesis and recruit immunosuppressive cells to the TME to evade the immune system [9].

While CAFs have historically been considered to be cancer-promoting components, recent studies have shown that CAFs could have tumor-restraining functions in certain circumstances [15]. The tumor-restraining actions of CAFs are likely dependent on stimulation of anticancer immunity, pro-inflammatory secretome, tumor inhibitory signaling, and the synthesis of ECM components as barriers to tumor cell invasion and dissemination [5]. A study in mice has shown that myofibroblast depletion leads to increased tumor invasion, which is associated with decreased survival [16]. This study suggests that CAFs have functions in restraining tumors. This paradoxical nature of CAFs can potentially be explained by the heterogeneity of CAFs [15].

2.2 Heterogeneity of CAFs

There is mounting evidence that CAFs are a heterogeneous population of cells [9]. This likely depends on the numerous precursors of CAFs [9]. CAFs can be recruited to the tumor from a distant source, such as bone marrow [14], or transdifferentiate from non-fibroblastic lineages, such as epithelial cells, blood vessels, adipocytes, pericytes, and smooth muscle cells [9]. Numerous precursors of CAFs are shown in Figure 3. The study of genetically modified mouse models (GEMMs) designed to limit the accumulation of CAFs in growing pancreatic tumors or to conditionally delete the vascular endothelial growth factors in breast CAFs revealed that there are distinct functional subtypes of CAFs [17].

2.3 Functions of CAFs

CAFs have both pro-tumor and antitumor tendencies [17]. Pro-tumorigenic functions of CAFs are generally driven by their altered secretive [17]. Paracrine signaling between cancer cells and CAFs leads to tumor progression by enhancing the survival, proliferation, stemness, and metastasis-initiating capacity of cancer cells, promoting cancer progression and enhancing resistance to therapy [17]. CAFs also have an indirect influence in promoting tumor growth due to their ability to remodel the ECM [17]. The stiffness of the tissue, which plays a critical role in tumorigenesis, is influenced by modifications in the ECM’s composition and cross-linking [18, 19]. CAFs express lysyl oxidase (LOX), an enzyme that cross-links and stiffens collagen fibers, promoting their stability [18]. CAFs also regulate the degradation of the ECM [18]. CAFs secrete cytokines and chemokines that regulate tumor immunity and the intratumoral vascular program [17]. Several studies have indicated that CAFs play an important role in chemoresistance via different mechanisms, including but not limited to increasing stem cancer cells, secreting cytokines, and secreting miRNAs [10]. miRNAs have been shown to inhibit tumor-repressor genes, thus promoting cell growth and invasion, metastasis, and tumorigenesis [20].

CAFs trigger tumor initiation and progression [18]. In vitro coculture and in vivo transplantation experiments have shown that human prostatic CAFs induced the proliferation and the ability to form tumors from immortalized nontumorigenic human prostatic epithelial cells [18]. This effect was not exhibited by normal fibroblasts. It is thought that CAFs’ secreted factors are what cause this tumor-initiating potential [18].

Numerous studies have shown that CAFs confer resistance to chemotherapy [6]. Some CAF-mediated resistant mechanisms include delivery of exosomes stimulating cancer cell survival, promoting cancer cell epithelial-mesenchymal transition, and thus decreasing expression of transporters responsible for drug uptake and scavenging chemo drug to reduce the amount of intratumoral chemotherapy drug [6]. CAFs also contribute to the resistance to targeted therapy [6]. Additionally, evidence suggests that CAFs contribute to immune evasion and immunotherapy resistance [6].

Antitumor functions of CAFs are predominantly associated with their functions as regulators of antitumor immunity [17]. Studies in mice have shown that fibroblast depletion leads to increased tumor invasion [16]. The use of defined gene promoter-driven expression of viral thymidine kinase proteins in GEMMs to study CAFs has allowed researchers to deplete populations of CAFs using ganciclovir, a substance that is toxic only to cells that express viral thymidine kinase [17]. A similar approach to deplete CAFs expressing αSMA suggested that αSMA+ stromal cells were predominantly acting to restrain cancer progression [17]. The depletion of these αSMA expressing CAFs yielded a more invasive tumor with enhanced intratumoral hypoxia [17]. A reduction in CAFs in GEMMs of pancreatic tumors with a deletion of sonic hedgehog (SHH) in the cancer cells also resulted in more aggressive tumors with increased cancer proliferation [17].

Advertisement

3. Targeting CAFs for cancer intervention

Numerous studies have proven CAFs’ significant role in cancer progression and subsequently the potential of CAFs as targets for effective cancer intervention. Traditionally, therapies involved targeting cancer cells directly [21]. Recent complementary efforts aim to disrupt the networks that promote cancer cell activity and behavior [21]. The depletion of CAFs and targeting of CAF-dependent pathways can indirectly result in malignant cell death through both immune-dependent and immune-independent mechanisms [21]. Most conventional cancer therapies, such as radiotherapy and chemotherapy, are likely to affect CAFs as well by preventing cellular division by inducing DNA damage, impeding DNA and RNA synthesis, and blocking the cytoskeleton remodel required for cell division [17]. However, the unintended impact of these therapeutic methods on the function and accumulation of CAFs is largely unknown [17].

As a result, research is being conducted to help target CAFs through alternative methods [21]. One approach involves targeting the regulatory pathways leading to fibroblast differentiation and activation [9, 17, 21]. For example, TGFβ is a common factor in the conversion of different cell types into CAFs. In a study, Mariathasan et al. found the two top scoring TGFβ pathway genes represent a ligand, TGFβ1, and receptor TGFβR2 [22]. In murine tumor models, blocking the TGFβ signaling by using the SM16 TGFβ receptor inhibitor or anti-TGFβ antibodies resulted in the recession of tumor growth [23]. By targeting these regulatory pathways, the activation of fibroblasts could be prevented, preventing CAFs from activating and functioning.

Another approach for targeting CAFs for cancer intervention is targeting CAF-secreted factors [11]. Numerous mitogens, chemokines, and matricellular proteins that CAFs release aid in the evolution of tumor progression and the development of drug resistance [11]. Targeting these CAF-secreted factors should prevent the promotion of tumor progression and drug resistance, making the tumor more susceptible to drugs. The heterogeneity of CAFs also proves as an advantage for cancer intervention via shifting the influence of pro- vs. antitumorigenic populations [21].

Currently, there are many drugs under trial as shown in Table 1. Of the potential targets identified in CAFs, fibroblast-activation protein (FAP) is the most studied. FAP has been neither detected in benign tumors nor in most normal quiescent adult stromal cells [35]. FAP is a type II integral membrane of the prolyl oligopeptidase family, or S9 family [36]. FAP is further classified into the dipeptidyl peptidase (DPP) subfamily (S9B) [36]. This class of enzymes is characterized by its capacity to cleave the pro-Xaa peptide link, where Xaa can be any amino acid. It has been demonstrated that this enzymatic activity contributes to the development of cancer by altering bioactive signaling peptides [36]. In vivo, FAP+ CAFs were successfully depleted by the FAP-depleting immunotoxin, and tumor models demonstrated strong tumor inhibitory effects. [6]. Other approaches in targeting FAP include DNA vaccine and chimeric antigen receptor (CAR) T cells [6].

DrugsTarget and mechanismCancer typesNational Clinical Trial numberStatusRef.
Sibrotuzumab131I-labeled anti-FAP mAbColorectal, non-small cell lung, breast, or head and neck cancersNCT02198274
NCT02209727
Phase I[24]
Calcipotriol ParicalcitolVitamin D analogueEarly-stage skin cancer, breast cancer, pancreatic cancerNCT03596073
NCT04617067
NCT02030860
NCT03138720
NCT04054362
Phase I/II[25]
Pamrevlumab (FG-3019)Anti-CTGF mAbPancreatic cancerNCT03941093Phase III[26]
Plerixafor (AMD3100) BL-8040 (motixafortide)CXCR4 receptor antagonistPancreatic cancerNCT04177810
NCT02179970
NCT02826486
NCT03193190
Phase I/II[27, 28]
IPI-926Smoothened inhibitorPancreatic cancerNCT01130142Phase I[29]
S-3304MMP inhibitorAdvanced solid tumorsNCT00078390
NCT00033215
Phase I[30]
131I-m81C6131I-labeled anti-tenascin mAbBrain tumorsNCT00002752
NCT00003461
Phase II[31]
ImatinibPDGFR inhibitorAdvanced solid tumorsNCT00161213
NCT00281996
NCT01048320
NCT00485485
Phase I/II[32]
GS-6624 (simtuzumab)LOXL2 mAbPancreatic cancerNCT01472198
NCT01479465
Phase II[33]
TetrathiomolybdateCopper
chelator, target LOX
Breast cancer, prostate cancerNCT00195091
NCT00150995
NCT00405574
Phase II[34]
Pegvorhyaluronidase alfa (PEGPH20; PVHA)Recombinant human hyaluronidaseLung cancer, pancreatic cancerNCT01453153
NCT02563548
NCT01839487
NCT02715804
Phase I/II/III

Table 1.

Summary of various drugs’ efficacy against CAF-induced cancer progression in clinical and pre-clinical studies [6].

Advertisement

4. Conclusions

Heterogeneous populations of CAFs exist in the TME. The heterogeneous nature of CAFs likely comes from their different origins, and this heterogeneity is likely the cause of the paradoxical nature of CAFs having both pro-tumorigenic and antitumorigenic functions. CAFs have many functions in the TME. CAFs alter the TME. They produce growth factors and pro-inflammatory cytokines.

CAFs are a promising target for cancer intervention. They have many pro-tumorigenic functions. CAFs can be targeted through their activation pathway by blocking a step in the pathway. One method is by preventing FAP from being produced by introducing siRNAs that are complementary to the FAP mRNA.

Over time, our understanding of CAFs and their contribution to cancer progression has expanded greatly. We now have a better understanding of their heterogeneity and their functions in the TME. While the antitumorigenic functions may act as a roadblock to targeting CAFs for cancer intervention, it may be possible to develop a treatment that targets the pro-tumorigenic functions of CAFs without targeting the antitumorigenic functions of CAFs by targeting subpopulations of CAFs that express pro-tumorigenic genes.

While CAFs are a promising target for cancer intervention due to their pro-tumorigenic functions, their antitumorigenic functions may act as a roadblock. Further research would be required before targeting CAFs as a conventional method of cancer intervention.

However, there are issues with targeting CAFs for cancer intervention. Figure 4 shows the effects of targeting only tumor cells vs. targeting only the TME on a tumor. In both, there is a possibility that the tumor can grow back. In addition, studies in mice have shown that fibroblast depletion leads to increased tumor invasion [16]. In mouse models, deletion of SHH accelerated the progression of pancreatic ductal adenocarcinoma [37].

Figure 4.

A schematic diagram of targeting tumor cell or TME only and their potential resistance mechanisms. Left: Targeting tumor cells only (such as chemotherapies) kills majority tumor cells. However, the residue tumor cells may survive due to the TME, leading to tumor relapse. Right: Targeting the TME can inhibit the recruitment and activation of pro-tumor cells and enhance antitumor responses. However, the TME will be reconstituted by tumor cells via recruitment and programming of bone marrow derived cells or local resident stromal or immune cells [6].

Advertisement

Acknowledgments

Funding for the publication of this paper was made possible by a grant from The Assisi Foundation of Memphis. Brown, Chester, PhD (PI). We also thank Yirui Tang for drawing the figures used in this paper.

Advertisement

Conflict of interest

The authors have declared that no conflict of interest exists.

References

  1. 1. Arneth B. Tumor microenvironment. Medicina. 2019;56(1):15
  2. 2. Soysal SD, Tzankov A, Muenst SE. Role of the tumor microenvironment in breast cancer. Pathobiology. 2015;82(3-4):142-152
  3. 3. Anderson NM, Simon MC. The tumor microenvironment. Current Biology. 2020;30(16):R921-R925. DOI: 10.1016/j.cub.2020.06.081 PMID: 32810447; PMCID: PMC8194051
  4. 4. Kirk T, Ahmed A, Rognoni E. Fibroblast memory in development, homeostasis and disease. Cell. 2021;10(11):2840
  5. 5. Chen Y, McAndrews KM, Kalluri R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nature Reviews Clinical Oncology. 2021;18(12):792-804
  6. 6. Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacology & Therapeutics. 2021;221:107753
  7. 7. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nature Reviews Cancer. 2020;20(3):174-186
  8. 8. Park D, Sahai E, Rullan A. SnapShot: Cancer-associated fibroblasts. Cell. 2020;181(2):486-486.e1. DOI: 10.1016/j.cell.2020.03.013 PMID: 32302576
  9. 9. Liu T, Han C, Wang S, Fang P, Ma Z, Xu L, et al. Cancer-associated fibroblasts: An emerging target of anti-cancer immunotherapy. Journal of Hematology & Oncology. 2019;12(1):1-15
  10. 10. Louault K, Li RR, DeClerck YA. Cancer-associated fibroblasts: Understanding their heterogeneity. Cancers. 2020;12(11):3108
  11. 11. Öhlund D, Elyada E, Tuveson D. Fibroblast heterogeneity in the cancer wound. Journal of Experimental Medicine. 2014;211(8):1503-1523
  12. 12. Tang PC, Chung JY, Xue VW, Xiao J, Meng XM, Huang XR, et al. Smad3 promotes Cancer-associated fibroblasts generation via macrophage-Myofibroblast transition. Advanced Science. 2022;9(1):e2101235. DOI: 10.1002/advs.202101235 PMID: 34791825; PMCID: PMC8728853
  13. 13. De P, Aske J, Dey N. Cancer-associated fibroblast functions as a road-block in Cancer therapy. Cancers. 2021;13(20):5246
  14. 14. Yoshida GJ. Regulation of heterogeneous cancer-associated fibroblasts: The molecular pathology of activated signaling pathways. Journal of Experimental & Clinical Cancer Research. 2020;39(1):1-15
  15. 15. Biffi G, Tuveson DA. Diversity and biology of cancer-associated fibroblasts. Physiological Reviews. 2021;101(1):147-176
  16. 16. Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, et al. Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25(6):719-734
  17. 17. LeBleu VS, Kalluri R. A peek into cancer-associated fibroblasts: Origins, functions and translational impact. Disease Models & Mechanisms. 2018;11(4):dmm029447
  18. 18. Pickup MW, Laklai H, Acerbi I, Owens P, Gorska AE, Chytil A, et al. Stromally derived Lysyl oxidase promotes metastasis of transforming growth factor-β–deficient mouse mammary CarcinomasStromal LOX promotes TGFβR2-null breast Cancer metastasis. Cancer Research. 2013;73(17):5336-5346
  19. 19. Cukierman, E., & Bassi, D. E. (2010). Physico-mechanical aspects of extracellular matrix influences on tumorigenic behaviors. In Seminars in cancer Biology (Vol. 20, No. 3, pp. 139-145). Academic Press.
  20. 20. Madu CO, Gu A, Madu C, Lu Y. The Role of MicroRNAs in the Progression, Prognostication, and Treatment of Breast Cancer. 2020
  21. 21. Barrett RL, Puré E. Cancer-associated fibroblasts and their influence on tumor immunity and immunotherapy. eLife. 2020;9:e57243
  22. 22. Mariathasan S et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature. 2018;554(7693):544-548. DOI: 10.1038/nature25501. Epub 2018 Feb 14
  23. 23. Zhao H et al. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduction and Targeted Therapy. 2021;6(1):263. DOI: 10.1038/s41392-021-00658-5
  24. 24. Siveke JT. Fibroblast-activating protein: Targeting the roots of the tumor microenvironment. Journal of Nuclear Medicine. 2018;59:1412-1414. DOI: 10.2967/jnumed.118.214361
  25. 25. Sherman MH et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell. 2014;159:80-93. DOI: 10.1016/j.cell.2014.08.007
  26. 26. Neesse A et al. CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:12325-12330. DOI: 10.1073/pnas.1300415110
  27. 27. Feig C et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:20212-20217. DOI: 10.1073/pnas.1320318110
  28. 28. Bockorny B et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: The COMBAT trial. Nature Medicine. 2020;26:878-+. DOI: 10.1038/s41591-020-0880-x
  29. 29. Jimeno A et al. Phase I study of the hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clinical Cancer Research. 2013;19:2766-2774. DOI: 10.1158/1078-0432.CCR-12-3654
  30. 30. Chiappori AA et al. A phase I pharmacokinetic and pharmacodynamic study of s-3304, a novel matrix metalloproteinase inhibitor, in patients with advanced and refractory solid tumors. Clinical Cancer Research. 2007;13:2091-2099. DOI: 10.1158/1078-0432.CCR-06-1586
  31. 31. Reardon DA et al. Salvage radioimmunotherapy with murine iodine-131-labeled antitenascin monoclonal antibody 81C6 for patients with recurrent primary and metastatic malignant brain tumors: Phase II study results. Journal of Clinical Oncology. 2006;24:115-122. DOI: 10.1200/JCO.2005.03.4082
  32. 32. Reilley MJ et al. Phase I clinical trial of combination imatinib and ipilimumab in patients with advanced malignancies. Journal for Immunotherapy of Cancer. 2017;5:35. DOI: 10.1186/s40425-017-0238-1
  33. 33. Benson AB 3rd et al. A phase II randomized, double-blind, placebo-controlled study of Simtuzumab or placebo in combination with gemcitabine for the first-line treatment of pancreatic adenocarcinoma. The Oncologist. 2017;22:e241-e215. DOI: 10.1634/theoncologist.2017-0024
  34. 34. Chan N et al. Influencing the tumor microenvironment: A phase II study of copper depletion using Tetrathiomolybdate in patients with breast Cancer at high risk for recurrence and in preclinical models of lung metastases. Clinical Cancer Research. 2017;23:666-676. DOI: 10.1158/1078-0432.CCR-16-13261078-0432.CCR-16-1326 [pii]
  35. 35. Wang LCS, Lo A, Scholler J, Sun J, Majumdar RS, Kapoor V, et al. Targeting fibroblast activation protein in tumor stroma with chimeric antigen receptor T cells can inhibit tumor growth and augment host immunity without severe ToxicityFAP-redirected CAR T cells. Cancer Immunology Research. 2014;2(2):154-166
  36. 36. Brennen WN, Isaacs JT, Denmeade SR. Rationale behind targeting fibroblast activation protein–expressing carcinoma-associated fibroblasts as a novel chemotherapeutic strategy. Molecular Cancer Therapeutics. 2012;11(2):257-266
  37. 37. Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25(6):735-747

Written By

Anyu Gu, Chikezie O. Madu and Yi Lu

Submitted: 27 August 2022 Reviewed: 31 October 2022 Published: 04 January 2023