Journal of Community Medicine & Public Health

Ambiguity of Cancer Associated Fibroblasts: Protumorigenic or Antitumorigenic

by Rupal Tripathi1, Anurag Mehta1,2*

1Department of Research, Rajiv Gandhi Cancer Institute & Research Centre, Delhi, India

2Department of Laboratory Services & Molecular Diagnostics, Rajiv Gandhi Cancer Institute & Research Centre, Delhi, India

*Corresponding author: Anurag Mehta, Director, Laboratory Services, Molecular Diagnostic & Research, Rajiv Gandhi Cancer Institute & Research Centre, Sector-5, Rohini, Delhi-110085, India

Received Date: 15 June, 2024

Accepted Date:01 July, 2024

Published Date:05 July, 2024

Citation: Tripathi R, Mehta A (2024) Ambiguity of Cancer Associated Fibroblasts: Protumorigenic or Antitumorigenic. J Community Med Public Health 8: 446. https://doi.org/10.29011/2577-2228.100446

Abstract

Tumor microenvironment today has at least equally if not more than the tumor itself succeeded in garnering attention due to its key roles in affecting the cancer cells by the non-mutant cells. The cell-to-cell interactions by itself play a major role in the development and progression of cancer. Cancer associated fibroblasts (CAFs), a key component of the tumor microenvironment have sparked interest due to their herculean task of modulating cancer metastasis, also including matrix deposition and remodelling and tumor mechanistics, even though, till today, a lacuna exists in the understanding. CAFs have also been known to affect drug access and therapy responses. A possibility of counteracting CAFs can also prove as an armor in the fight against the dreaded disease cancer. However, there is an underlying hurdle as it is yet to be established whether its protumorigenic capabilities weigh down the antitumorigenic effects. This delinquent situation is largely affected by the inability to find the specific markers of these cell types. The present review deals with the identification, generation, functionality and challenges associated with the use of CAFs in depth.

Keywords: Tumor microenvironment; Cancer associated fibroblasts; Fibroblasts; Cancer

Introduction to CAFs

The tumor microenvironment is a multicellular system in which the cells in the extracellular matrix interact closely with the tumor cells [1,2]. This cross-talk may be having either a positive or a negative stimulus. These cells are mainly from the mesenchyme, endothelium, and hematopoietic origin influencing the process of tumorigenesis. One of the major components in the stromal environment are the CAFs. Conventionally, cells presenting with an elongated morphology and having a negative expression for epithelial, endothelial, and leukocyte markers qualify as CAFs. This is further potentiated by the lack of mutations found in the cancer cells [1]. However, cancer cells undergoing Epithelialto-Mesenchymal Transition (EMT) have been excluded from this category. In the Banbury Center meeting at Cold Spring Harbor Laboratory, New York, USA in 2019, the experts discussed the current understanding of CAF biology and looked into the fundamental properties of CAFs and its applications [1]. Fibroblasts are generally quiescent which get activated in a wound-healing response and CAFs can be effectively put to use in anti-cancer immunotherapy [3-5]. It remains to be ascertained as to whether the common and specific traits of CAFs have been preserved across generations and lineages [6-8].

Origin, Evolution and Generation

The primitive mesenchyme developed from the mesoderm after the process of gastrulation along with a subset of fibroblasts derived from the neural crest, as part of the ectoderm [9]. There is mounting evidence which shows that the origin of CAFs lies in the resident fibroblasts, dedifferentiating mature cells, and also from the tumor cells [10]. This adds to the woes in establishing and characterizing the fibroblasts which are recognized on the basis of their morphology and position and lack of markers like epithelial cells, endothelial cells and leukocytes. However, markers for its subtypes are typically being used including fibroblast activation protein (FAP) and alpha-smooth muscle actin (alpha SMA) [1112]. Alpha SMA is typically expressed when the TGF beta through SMAD-dependent and independent pathways activates fibroblasts into CAFs [13]. Another important player secreted from the stromal and tumor cells is the transforming growth factor beta 1 (TGF- beta1) It has also been documented that the fibroblasts affect the local epithelial stem cell behavior, promote angiogenesis and harmonize the functioning of the immune system thereby promoting immune tolerance and also maintain the metabolic homeostasis. This prompts us in establishing the diverse roles of fibroblasts during key processes like normal tissue homeostasis and repair [14-19]. Additionally, CAFs can also be derived from the mesenchymal stromal cells expressing markers like alpha SMA.

In order to partially counter the problem, laboratories are increasingly and efficiently looking into the step-by-step changes leading to the formation of a cancer cell from a normal healthy cell and typically, the fibroblastic components involved in the transformation. In this context, the concept of stromatogenesis seems to have evolved in line with tumorigenesis. It is believed that the malignant transformation is also accompanied by the expansion of stromal fibroblasts. The varied areas of CAFs is being extensively studied with reference to the entire cancer spectrum. Figure 1 shows the pathology of CAFs in the different cancers.

 

Figure 1: Representative images of cancer associated fibroblasts (CAFs) in (a) pancreas (b) breast.

Mechanisms of Activation

The routine physiological roles played by fibroblasts greatly impact the key behavior and functioning of the CAFs. The normal fibroblasts can lead to the activation of CAFs by a variety of mechanisms as shown in Figure 2. These include the contact signals (Notch and Eph-ephrins), extracellular matrix (stiffness and composition), DNA damage (chemotherapy and radiotherapy), Transforming Growth Factor- beta (TGF- beta), physiological stress (reactive oxygen species and disrupted metabolism), inflammatory signals [interleukins (IL) 1 & 6, Tumour Necrosis Factor (TNF), Receptor Tyrosine Kinase (RTK) ligands [platelet derived growth factor (PDGF) and Fibroblast Growth Factor (FGF). TGF- beta drives the expression of alpha SMA and increase the activity of contractile cytoskeletons [12,20-22]. In pancreatic cancer, the proliferation of CAFs is increased by vitamins A and D which increase the activation of CAFs into a pro-tumorigenic state [16,23,24]. This is achieved typically by the SMAD signaling pathway or the renin-angiotensin mechanism. Double-stranded breaks in DNA can stimulate the production of IL-6 and the TGFβ family ligand activin A [25,26]. Activation of signaling pathways including NF kappa B and ERK pathways help in the generation of CAFs from the resident fibroblasts along with the other factors secreted from the tumor cells [27-30]. Inflammatory modulators like interleukins (typically 1 and 6) also promote the activation of CAF working alongside NF kappa B and signal transducer and activator of STAT transcription factors [31,32]. CAF activation is also prompted by the involvement of JAK-STAT signaling and alterations in chromatin modification typically histone acetylation alterations [33,34]. Generation of reactive oxygen species and activation of hypoxia inducible factor (HIF 1alpha) also drive the activation of resident fibroblasts to CAFs [35-37].

Key Roles

Generation of CAFs and in turn its functioning is greatly dependent on its origin. Overall, they present as a very heterogenous population of cells [38]. A clear definition of CAF subpopulations and linkage with their functionality till today remains a challenge, chiefly due to the lack of specific biological markers. CAFs generated from the tumor itself or the tumor microenvironment in turn affect its functionality as being pro or ani-tumorigenic (Figure 2). In general, cancer therapies are also known to initiate the generation of CAFs which is an anti-tumorigenic property of CAFs. However, the catch remains that it can lead to therapy resistance. CAFs play a significant role in matrix remodeling which typically includes matrix crosslinking, proteolysis, and matrix production and remodeling. It also mediated the immune crosstalk by activation of TGF beta, IL-6 production, CXCL12, and CCL2 production. Metabolic effects which included lactate, alanine, and aspartate shuttling and amino acid depletion, and soluble secreted factors including VEGF, exosomes, and HGF and GAS6 production are also carried out by the CAFs. Matrix remodeling and immune crosstalk in turn regulate the cancer

cell invasion and the interference with T cell functioning, respectively. Macrophage and endothelial crosstalk are also delimited by the soluble secreted factors. Metabolic effects on the other hand, greatly regulate the growth of the tumor cells. CAFs also secrete large volumes of growth factors, cytokines, and exosomes. The functional heterogeneity of the CAFs has been greatly identified and defined in a variety of cancers. In the cancers of the breast and pancreas, CAFs are typically associated with angiogenesis, metastasis, immunosuppression, angiogenesis, migration, invasion, and chemoresistance whereas in oral squamous cell carcinoma, it is typically associated with invasion, immunosuppression, and migration [39-44].

 

Figure 2: Mechanisms of activation of CAFs.

Also, in contrast to the pro-tumorigenic functions, CAFs are also involved in anti-tumorogenicity. An inhibition of factors including VEGF, CXCL-12, and IL-8 has been observed, hence inhibiting pathways affecting the growth of tumors, immunosuppression, and angiogenesis [45,46]. This is typically achieved by increased activation of Shh signaling by CAFs expressing alpha SMA. An inhibition of tumor cell proliferation has also been reported in prostatic adenocarcinomas and sarcomas due to increased production of TGF beta, and Tumor necrosis factor alpha [47-50]. In tumors of the intestine, tumor growth and angiogenesis is checked by NF kappa B signaling in CAFs [51].

Challenges

Despite the benefits of the CAFs as reported in the trials, the key challenge still remains with its mode of usage in combating cancer. One way can be by using strategies to directly target the CAFs and eliminating them. There still remains a dearth of markers in identifying the CAFs and hence the delay exists in eliminating them. A protein, alpha FAP which is expressed in greater than 75% CAFs has been routinely targeted [52]. Chimeric antigen receptor T cells and monoclonal antibodies are being explored in tumors of breast, lung and colon. CAR-T cells are particularly being looked into with the advantage of increased efficacy and little/ no toxicity. Growth factors including platelet derived and tumor growth factors have been widely linked to the activation of CAFs. Its activation has also been linked to the epigenetic changes involving regulatory miRNAs. The identification of these strategies further provides the opportunity and evidence to directly target these CAFs [53]. Apart from this strategy, another way of targeting CAFs can be done by targeting the proteins secreted by CAFs or also by using the CAFs to deliver the anti-tumor molecules. Table 1 lists the drugs involved and their underlying mechanisms in directly targeting the CAFs.

Name of drug

Phase of trial

Cancer site

Mechanism

Reference

Sibrotuzumab

I

NSCLC, Melanoma, Pancreas

Monoclonal antibody against alphaFAP

[54-56]

TRC-105

II

Neuroblastoma

Monoclonal antibody against endoglin

[57]

Calcipotriol

II

Pancreas

Vitamin D receptor activation and stellate cell deactivation

[58]

Ruxolitinib

I & II

Pancreas, Lung

JAK inhibitor

[59-61]

Losartan

II

Pancreas, Melanoma

Angiotensin receptors blocker

[62,63]

Bevacizumab, FOLFIRINOX

II

Pancreas, Colorectum

Neutralization of VEGF

[64,65]

Abraxane, Gemcitabine

II

Pancreas

Decreases type I collagen production and secretion of CXCL10 and IL6

[60]

Nab-paclitaxel, Atezolizumab

III

Breast, Lung

Monoclonal antibody against PDL1

[66-68]

Plerixafor

Murine

model

Prostate, Pancreas

Small molecule inhibitor for CXCR4

[69-71]

Galunisertib

Ib/ II

Pancreas

Small molecule targeting TGF beta receptor

[72]

Defactinib

I/II

Pancreas, Ovary

Small molecule inhibitor reducing signaling downstream of integrins

[73]

Saridegib, Vismodegib

I/II

Breast, Prostate, Ovary, Lung, Pancreas, Stomach, etc.

Small molecule inhibitor reducing CAF activation

[74,75]

Dasatinib

I/II

Breast, Prostate, Ovarian, Endometrium

Tyrosine kinase inhibitors

[76]

Tasisulam

I/II

Breast, Ovarian, Lung, Kidney

TGF beta receptor kinase inhibitors

[76]

Crenolanib

I/II

Gastrointestinal tumors

PDGFR tyrosine kinase inhibitors

[76]

Other drugs e.g. curcumin

I/II

Breast, Prostate, Lung, Head and Neck, Brain, etc.

Miscellaneous

[76]

NSCLC: Non-Small Cell Lung Cancer; FAP: Fibroblast Activation Protein; JAK: Janus Kinase; VEGF: Vascular Endothelial Growth Factor; CXCL: Chemokine (C-X-C motif) Ligand; IL: Interleukin; PDL1: Programmed Cell Death Ligand 1; CXCR: C-X-C Chemokine Receptor; TGF:

Transforming Growth Factor; CAF: Cancer Associated Fibroblasts; PDGFR: Platelet-Derived Growth Factor Receptor

Table 1: Drugs involved in directly targeting the cancer associated fibroblasts.

Points to ponder

In the times to come, both the pro and anti-tumorigenic properties of the CAFs can be fully utilized for their use in cancer-directed therapies, especially with the improved understanding of their heterogeneity. Also, targeting CAFs and their secretome can also greatly help in targeting the tumor microenvironment without directly affecting the tumor cells as the tumors are residing in a typically fibroblast-rich microenvironment. Clinical trials evaluating the same should be very careful and vigilant in reporting the results especially taking into consideration the morphology, pathology, and functionality of these fibroblasts. How these fibroblasts depict themselves greatly drives the functionality and their possible usage in future therapies. A very comprehensive and robust nomenclature capturing these findings

should be adopted and put into practice universally and evolving a common nomenclature across the cancer types can greatly give the required impetus in this area. Spatial transcriptomics can be effectively put to use in order to address the problem adequately by integrating histopathological annotation and high-throughput sequencing in a unified framework with the objective of identifying the phenotypic heterogeneity and functional diversity of CAFs.

Statements and Declarations Competing Interests: None to disclose

Author Contributions

RT: Conceptualization, Writing the review

AM: Conceptualization, Finalizing the review

References

  1. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, et al. (2020) A framework for advancing our understanding of cancerassociated fibroblasts. Nat Rev Cancer 20: 174-186.
  2. Maman S, Witz IP (2018) A history of exploring cancer in context. Nat Rev Cancer 18: 359-376.
  3. Gajewski TF, Schreiber H, Fu YX (2013) Innate and adaptive immune cells in the tumor microenvironment. Nat Immunol 14: 1014-1022.
  4. Petty AJ, Yang Y (2017) Tumor-associated macrophages: implications in cancer immunotherapy. Immunotherapy 9: 289-302.
  5. Zhang D, Zheng Y, Lin Z, Liu X, Li J, et al. (2020) Equipping Natural Killer Cells with Specific Targeting and Checkpoint Blocking for Enhanced Adoptive Immunotherapy in Solid Tumors. Angew Chem Int Ed Engl 59: 12022-12028.
  6. Puram SV, Tirosh I, Parikh AS, Patel AP, Yizhak K, et al. (2017) Single- cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171: 1611-1624.
  7. Croft AP, Campos J, Jansen K, Turner JD, Marshall J, et al. (2019) Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 570: 246-251.
  8. Nayar S, Campos J, Smith CG, Iannizzotto V, Gardner DH, et al. (2019) Immunofibroblasts are pivotal drivers of tertiary lymphoid structure formation and local pathology. Proc Natl Acad Sci USA 116: 13490-13497.
  9. Sharpe PT (2001) Neural crest and tooth morphogenesis. Adv Dent Res 15: 4-7.
  10. Haviv I, Polyak K, Qiu W, Hu M, Campbell I (2009) Origin of carcinoma associated fibroblasts. Cell Cycle 8: 589-595.
  11. Roberts EW, Deonarine A, Jones JO, Denton AE, Feig C, et al. (2013) Depletion of stromal cells expressing fibroblast activation protein- alpha from skeletal muscle and bone marrow results in cachexia and anemia. J Exp Med 210: 1137-1151.
  12. Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA (2002) Myofibroblasts and mechano- regulation of connective tissue remodelling. Nat Rev Mol Cell Biol 3: 349-363.
  13. Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, et al. (2004) TGF- Beta Signaling in Fibroblasts Modulates the Oncogenic Potential of Adjacent Epithelia. Science 303: 848-851.
  14. Brown FD, Turley SJ (2015) Fibroblastic reticular cells: organization and regulation of the T lymphocyte life cycle. J Immunol 194: 13891394.
  15. Fletcher AL, Malhotra D, Turley SJ (2011) Lymph node stroma broaden the peripheral tolerance paradigm. Trends Immunol 32: 12-18.
  16. Sherman MH, Yu RT, Engle DD, Ding N, Atkins AR, et al. (2014) Vitamin D receptor- mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 159: 80-93.
  17. Apte M, Pirola RC, Wilson JS (2015) Pancreatic stellate cell: physiologic role, role in fibrosis and cancer. Curr Opin Gastroenterol 31: 416-423.
  18. Blaner WS, O’Byrne SM, Wongsiriroj N, Kluwe J, D’Ambrosio DM, et al. (2009) Hepatic stellate cell lipid droplets: a specialized lipid droplet for retinoid storage. Biochim Biophys Acta 1791: 467-473.
  19. Sherman MH, Yu RT, Tseng TW, Sousa CM, Liu S, et al. (2017) Stromal cues regulate the pancreatic cancer epigenome and metabolome. Proc Natl Acad Sci USA 114: 1129-1134.
  20. De Wever O, Nguyen Q, Van Hoorde L, Bracke M, Bruyneel E, et al. (2004) Tenascin- C and SF/HGF produced by myofibroblasts in vitro provide convergent proinvasive signals to human colon cancer cells through RhoA and Rac. FASEB J 18: 1016-1018.
  21. Calvo F, Ege N, Grande-Garcia A, Hooper S, Jenkins RP, et al. (2013) Mechanotransduction and YAP- dependent matrix remodelling is required for the generation and maintenance of cancer- associated fibroblasts. Nat Cell Biol 15: 637-646.
  22. Foster CT, Gualdrini F, Treisman R (2017) Mutual dependence of the MRTF- SRF and YAP- TEAD pathways in cancer- associated fibroblasts is indirect and mediated by cytoskeletal dynamics. Genes Dev 31:  2361-2375.
  23. Liu T, Han C, Wang S, Fang P, Ma Z, et al. (2019) Cancer-associated fibroblasts: an emerging target of anti-cancer immunotherapy. J Hematol Oncol 12: 86.
  24. Shany S, Sigal-Batikoff I, Lamprecht S (2016) Vitamin D and Myofibroblasts in Fibrosis and Cancer: At Cross-purposes with TGF-/ SMAD Signaling. Anticancer Res 36: 6225-6234.
  25. Fordyce CA, Patten KT, Fessenden TB, DeFilippis RA, Hwang ES, et al. (2012) Cell- extrinsic consequences of epithelial stress: activation of protumorigenic tissue phenotypes. Breast Cancer Res 14: R155.
  26. Fordyce C, Fessenden T, Pickering C, Jung J, Singla V, et al. (2010) DNA damage drives an activin a- dependent induction of cyclooxygenase-2 in premalignant cells and lesions. Cancer Prev Res 3: 190-201.
  27. Tejada ML, Yu L, Dong J, Jung K, Meng G, et al. (2006) Tumor-driven paracrine platelet-derived growth factor receptor alpha signaling is a key determinant of stromal cell recruitment in a model of human lung carcinoma. Clin Cancer Res 12: 2676-2688.
  28. Erez N, Truitt M, Olson P, Arron ST, Hanahan D (2010) CancerAssociated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaBDependent Manner. Cancer Cell 17: 135-147.
  29. Tian H, Callahan CA, DuPree KJ, Darbonne WC, Ahn CP, et al. (2009) Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc Natl Acad Sci USA 106: 4254-4259.
  30. Elenbaas B, Weinberg RA (2001) Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation. Exp Cell Res 264: 169-184.
  31. Erez N, Truitt M, Olson P, Arron ST, Hanahan D (2010) Cancer- associated fibroblasts are activated in incipient neoplasia to orchestrate tumorpromoting inflammation in an NF- kappaB-dependent manner. Cancer Cell 17: 135-147.
  32. Sanz-Moreno V, Gaggioli C, Yeo M, Albrengues J, Wallberg F, et al. (2011) ROCK and JAK1 signaling cooperate to control actomyosin contractility in tumor cells and stroma. Cancer Cell 20: 229-245.
  33. Albrengues J, Bertero T, Grasset E, Bonan S, Maiel M, et al. (2015) Epigenetic switch drives the conversion of fibroblasts into proinvasive cancerassociated fibroblasts. Nat Commun 6: 10204.
  34. Albrengues J, Bourget I, Pons C, Butet V, Hofman P, et al. (2014) LIF mediates proinvasive activation of stromal fibroblasts in cancer. Cell Rep 7: 1664-1678.
  35. Costa A, Scholer-Dahirel A, Mechta-Grigoriou F (2014) The role of reactive oxygen species and metabolism on cancer cells and their microenvironment. Semin Cancer Biol 25: 23-32.
  36. Zou B, Liu X, Zhang B, Gong Y, Cai C, et al. (2018) The Expression of FAP in Hepatocellular Carcinoma Cells is Induced by Hypoxia and Correlates with Poor Clinical Outcomes. J Cancer 9: 3278-3286.
  37. Radisky DC, Levy DD, Littlepage LE, Liu H, Nelson CM, et al. (2005) Rac1b and Reactive Oxygen Species Mediate MMP-3-Induced EMT and Genomic Instability. Nature 436: 123-127.
  38. Louault K, Li RR, DeClerck YA (2020) Cancer-Associated Fibroblasts: Understanding Their Heterogeneity. Cancers (Basel) 12: 3108.
  39. Sugimoto H, Mundel TM, Kieran MW, Kalluri R (2006) Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther 5: 1640-1646.
  40. O’Connell JT, Sugimoto H, Cooke VG, MacDonald BA, Mehta AI, et al. (2011) VEGF-A and Tenascin-C Produced by S100A4+ Stromal Cells Are Important for Metastatic Colonization. Proc Natl Acad Sci USA 108: 16002-16007.
  41. Jiao J, González Á, Stevenson HL, Gagea M, Sugimoto H, et al. (2018) Depletion of S100A4+ Stromal Cells Does Not Prevent HCC Development but Reduces the Stem Cell-Like Phenotype of the Tumors. Exp Mol Med 50: e422.
  42. Carstens JL, Correa de Sampaio P, Yang D, Barua S, Wang H, et al. (2017) Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer. Nat Commun 8: 15095.
  43. Costea DE, Hills A, Osman AH, Thurlow J, Kalna G, et al. (2013) Identification of two distinct carcinoma-associated fibroblast subtypes with differential tumor-promoting abilities in oral squamous cell carcinoma. Cancer Res 73: 3888-3901.
  44. Hassona Y, Cirillo N, Heesom K, Parkinson EK, Prime SS (2014) Senescent Cancer-Associated Fibroblasts Secrete Active MMP-2 That Promotes Keratinocyte Dis-Cohesion and Invasion. Br J Cancer 111: 1230-1237.
  45. Özdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu C-C, et al. (2014) Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell 25: 719-734.
  46. Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, et al. (2014) Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell 25: 735-747.
  47. Alkasalias T, Flaberg E, Kashuba V, Alexeyenko A, Pavlova, T, et al. (2014) Inhibition of tumor cell proliferation and motility by fibroblasts is both contact and soluble factor dependent. Proc Natl Acad Sci USA 111: 17188-17193.
  48. Alexeyenko A, Alkasalias T, Pavlova T, Szekely L, Kashuba V, et al. (2015) Confrontation of fibroblasts with cancer cells in vitro: gene network analysis of transcriptome changes and differential capacity to inhibit tumor growth. J Exp Clin Cancer Res 34: 62.
  49. Degeorges A, Tatoud R, Fauvel-Lafeve F, Podgorniak MP, Millot G, et al. (1996) Stromal cells from human benign prostate hyperplasia produce a growth-inhibitory factor for LNCaP prostate cancer cells, identified as interleukin-6. Int J Cancer 68: 207-214.
  50. Paland N, Kamer I, Kogan-Sakin I, Madar S, Goldfinger N, et al. (2009) Differential influence of normal and cancer-associated fibroblasts on the growth of human epithelial cells in an in vitro cocultivation model of prostate cancer. Mol Cancer Res 7: 1212-1223.
  51. Pallangyo CK, Ziegler PK, Greten FR (2015) IKKβ acts as a tumor suppressor in cancer-associated fibroblasts during intestinaltumorigenesis. J Exp Med 212: 2253-2266.
  52. Wen Y, Wang CT, Ma TT, Li ZY, Zhou LN, et al. (2010) Immunotherapy targeting fibroblast activation protein inhibits tumor growth and increases survival in a murine colon cancer model. Cancer Sci 101: 2325-2332.
  53. Scott AM, Wiseman G, Welt S, Adjei A, Lee FT, et al. (2003) A Phase I dose-escalation study of sibrotuzumab in patients with advanced or metastatic fibroblast activation protein-positive cancer. Clin Cancer Res 9: 1639-1647.
  54. Hofheinz RD, al-Batran SE, Hartmann F, Hartung G, Jäger D, et al. (2003) Stromal antigen targeting by a humanised monoclonal antibody: an early phase II trial of sibrotuzumab in patients with metastatic colorectal cancer. Onkologie 26: 44-48.
  55. Narra K, Mullins SR, Lee HO, Strzemkowski-Brun B, Magalong K, et al. (2007) Phase II trial of single agent Val-boroPro (Talabostat) inhibiting Fibroblast Activation Protein in patients with metastatic colorectal cancer. Cancer Biol Ther 6: 1691-1699.
  56. Wu HW, Sheard MA, Malvar J, Fernandez GE, DeClerck YA, et al. (2019) Anti-CD105 Antibody Eliminates Tumor Microenvironment Cells and Enhances Anti-GD2 Antibody Immunotherapy of Neuroblastoma with Activated Natural Killer Cells. Clin Cancer Res 25: 4761-4774.
  57. Kobayashi H, Enomoto A, Woods SL, Burt AD, Takahashi M, et al. (2019) Cancer-associated fibroblasts in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol 16: 282-295.
  58. Hurwitz HI, Uppal N, Wagner SA, Bendell JC, Beck JT, et al. (2015) Randomized, Double-Blind, Phase II Study of Ruxolitinib or Placebo in Combination with Capecitabine in Patients With Metastatic Pancreatic Cancer for Whom Therapy With Gemcitabine Has Failed. J Clin Oncol 33: 4039-4047.
  59. Park JS, Hong MH, Chun YJ, Kim HR, Cho BC (2019) A phase Ib study of the combination of afatinib and ruxolitinib in EGFR mutant NSCLC with progression on EGFR-TKIs. Lung Cancer 134: 46-51.
  60. Yu HA, Perez L, Chang Q, Gao SP, Kris MG, et al. (2017) A Phase 1/2 Trial of Ruxolitinib and Erlotinib in Patients with EGFR-Mutant Lung Adenocarcinomas with Acquired Resistance to Erlotinib. J. Thorac Oncol 12: 102-109.
  61. Diop-Frimpong B, Chauhan VP, Krane S, Boucher Y, Jain RK (2011) Losartan inhibits collagen I synthesis and improves the distribution and efficacy of nanotherapeutics in tumors. Proc Natl Acad Sci USA 108: 2909-2914.
  62. Masamune A, Hamada S, Kikuta K, Takikawa T, Miura S, et al. (2013) The angiotensin II type I receptor blocker olmesartan inhibits the growth of pancreatic cancer by targeting stellate cell activities in mice. Scand J Gastroenterol 48: 602-609.
  63. Osumi H, Matsusaka S, Wakatsuki T, Suenaga M, Shinozaki E, et al. (2015) Angiotensin II type-1 receptor blockers enhance the effects of bevacizumab-based chemotherapy in metastatic colorectal cancer patients. Mol Clin Oncol 3: 1295-1300.
  64. Murphy JE, Wo JY, Ryan DP, Clark JW, Jiang W, et al. (2019) Total Neoadjuvant Therapy with FOLFIRINOX in Combination With Losartan Followed by Chemoradiotherapy for Locally Advanced Pancreatic Cancer: A Phase 2 Clinical Trial. JAMA Oncol 5: 1020-1027.
  65. Schmid P, Rugo HS, Adams S, Schneeweiss A, Barrios CH, et al. (2020) Atezolizumab plus nab-paclitaxel as first-line treatment for unresectable, locally advanced or metastatic triple-negative breast cancer (IMpassion130): updated efficacy results from a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 21: 4459.
  66. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, et al. (2018) Atezolizumab and Nab-Paclitaxel in Advanced Triple-Negative Breast Cancer. N Engl J Med 379: 2108-2121.
  67. West H, McCleod M, Hussein M, Morabito A, Rittmeyer A, et al. (2019) Atezolizumab in combination with carboplatin plus nab-paclitaxel chemotherapy compared with chemotherapy alone as first-line treatment for metastatic non-squamous non-small-cell lung cancer (IMpower130): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol 20: 924-937.
  68. Feig C, Jones JO, Kraman M, Wells RJB, Deonarine A, et al. (2013) Targeting CXCL12 from FAP- Expressing Carcinoma-Associated Fibroblasts Synergizes with Anti-PD-L1 Immunotherapy in Pancreatic Cancer. Proc Natl Acad Sci USA 110: 20212-20217.
  69. Zhu WB, Zhao ZF, Zhou X (2019) AMD3100 inhibits epithelialmesenchymal transition, cell invasion, and metastasis in the liver and the lung through blocking the SDF-1α/CXCR4 signaling pathway in prostate cancer. J Cell Physiol 234: 11746-11759.
  70. Daniel SK, Seo YD, Pillarisetty VG (2019) The CXCL12-CXCR4/ CXCR7 axis as a mechanism of immune resistance in gastrointestinal malignancies. Semin Cancer Biol 65: 176-188.
  71. Melisi D, Garcia-Carbonero R, Macarulla T, Pezet D, Deplanque G, et al. (2019) TGFβ receptor inhibitor galunisertib is linked to inflammation- and remodeling-related proteins in patients with pancreatic cancer. Cancer Chemother Pharmacol 83: 975-991.
  72. U.S. National Library of Medicine (2017) Phase II Study of VS-6063 in Patients with KRAS Mutant Non-Small Cell Lung Cancer. ClinicalTrials. gov.
  73. U.S. National Library of Medicine (2017) A Study Evaluating IPI-926 in Combination with Gemcitabine in Patients with Metastatic Pancreatic Cancer. ClinicalTrials.gov.
  74. U.S. National Library of Medicine (2017) Vismodegib and Gemcitabine Hydrochloride in Treating Patients With Advanced Pancreatic Cancer. ClinicalTrials.gov.
  75. Gascard P, Tlsty TD (2016) Carcinoma-associated fibroblasts: orchestrating the composition of malignancy. Genes Dev 30: 10021019.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. Read More About Open Access Policy.

Update cookies preferences