Another important component of the TME are the immune cells. Pioneering work by James Alisson
[108] and Tasuko Honjo
[109][110][111][109,110,111] in immune regulation has revolutionised the field of cancer immunology and established the groundwork for the development of numerous immunotherapies. Their respective findings that CTLA-4 and PD1 immune checkpoints inhibit the activity of cytotoxic T-cells and allow tumours to grow, led to the design of inhibitors against these molecules with the goal to enhance T-cell-mediated cell death of tumour cells
[112]. These checkpoint inhibitors showed remarkable results, especially in melanoma patients
[113][114][115][113,114,115]. However, it is not fully understood what factors dictate their efficiency and the durability of the patients’ response to these therapies. In addition to checkpoint inhibitors, several other immunotherapies are now available, including cancer vaccines
[116][117][116,117] and the adoptive transfer of immune cells, such as chimeric antigen receptor (CAR) T-cells
[118][119][118,119], and oncolytic viruses
[120]. Due to the strong influence that fibroblasts can exert in the immune milieu of the TME
[68][121][122][68,121,122], this cell type has emerged as a key modulator of the outcome of patients to these therapies. The aforementioned immunosuppressive molecules secreted by fibroblasts, namely TGF-β, IL-6, IL-8, CXCL12, can inhibit cytotoxic T-cell activity
[123][124][125][126][123,124,125,126], drive myeloid-derived suppressor cell (MDSC) differentiation
[127][128][129][127,128,129], modulate the phenotype of macrophages
[130][131][132][133][130,131,132,133], promote Treg formation
[54][134][54,134] and regulate the activity of natural killer (NK) cells
[132][135][132,135]. In addition to these, further studies have described other mechanisms through which fibroblasts regulate the immune landscape in tumours. α-SMA+ CAF (myCAFs) also secrete metabolic reprogramming factors, such as IDO1, Arg2 and galectin, which are responsible for generating an immunosuppressive TME via inducing T cell anergy and inhibiting CD8
+ T cell proliferation
[136]. The production and deposition of ECM proteins by CAFs strongly regulate the T-cell location within the tumours
[137]. A dense stroma can result in the exclusion of lymphocytes from areas rich in tumour cells, which results in poor adaptive immunity against the tumour
[138][139][138,139]. Moreover, the production of certain ECM proteins by fibroblasts, namely tenascin C or thrombospondin 1 (TSP1), can negatively impact the adhesion of T-cells
[140] and their activity
[141] in the TME, respectively. Fibroblasts can express immune checkpoint molecules themselves, such as PD-L1
[142], PD-L2
[37][142][37,142] and B7H3
[37][143][144][37,143,144], which can all inhibit T-cell activation. The production of CCL5 by stromal fibroblasts leads to an immunosuppressive environment as a result of the recruitment of Treg cells into the TME
[145]. The secretion of PGE
2, which was recently shown to characterise the apCAF subpopulation, can also result in the expansion of regulatory T-cells
[39]. Furthermore, PGE
2 is capable of inhibiting NK cell function
[146][147][148][146,147,148].
Fibroblasts not only communicate with the immune system via secreted factors, but they can also directly interact with CD8
+ T-cells. The HLA-class I antigen-presentation by stromal fibroblasts along with the expression of PD-L2 and FASL results in the killing of antigen-specific cytotoxic T-cells
[149]. Direct interaction between stromal fibroblasts and cancer cells also seems to drive resistance to oncolytic viruses as a result of the induction of a STING/IRF3-dependent inflammatory program in fibroblasts, which upregulates IFNβ1. The secretion of IFNβ1 into the TME induces an IFN-transcriptional program in cancer cells, rendering them less sensitive to infection by oncolytic viruses
[150].
2.3. Impact of Anti-Cancer Therapies in Fibroblasts
Further evidence that supports the study and detailed investigation of fibroblast dynamics during therapy is that stromal populations are strongly modulated and influenced by a wide variety of treatments applied in the clinics, ranging from cytotoxic agents to targeted agents
[151][152][153][151,152,153].
2.3.1. Direct Effects on CAFs
Most cytotoxic agents, such as chemotherapy and radiotherapy, lead to extensive DNA damage, which can result in cell cycle arrest and, consequently, senescence
[154][155][154,155]. Senescence is associated with the secretion of a vast array of cytokines, chemokines and growth factors, such as CCL2, IL-6, VEGF and TGF-β, which can aid cancer cell survival and, thus, contribute to therapeutic failure
[155][156][157][155,156,157]. Chan et al. showed that high doses of chemotherapy induce a potent remodelling of fibroblasts by activation of the JAK-STAT1 and NF-κB pathways, which results in the upregulation and secretion of several chemokines that, in their turn, support tumour-initiating cells and lead to chemoresistance
[82]. Similarly, another study described an increased risk of developing resistance to chemotherapy after treatment with the maximum tolerated dose due to drug-induced changes in the tumour stroma
[158]. In both cases, the effects observed in the tumour stroma could be prevented by adopting a metronomic (low-dose) chemotherapy regimen
[82][158][82,158]. In general, chemotherapy seems to activate an inflammatory gene signature in stromal fibroblasts, which is associated with a pro-tumorigenic state
[159]. The activation of the NF-κB signalling pathway by therapy-induced damage also promoted chemoresistance by driving the expression and secretion of WNT16B by fibroblasts and the subsequent activation of the Wnt program in cancer cells
[160]. The secretion of interleukins (ILs) by CAFs has been reported after treatment with chemotherapy. For example, IL-17A derived from chemotherapy-treated CAFs led to the expansion of tumour-initiating cells and, consequently, was shown to contribute to therapeutic failure
[63]. The exposure of fibroblasts to chemotherapy also resulted in a higher secretion of IL-11, which activated STAT3 signalling in cancer cells and drove the upregulation of anti-apoptotic pathways
[161]. In addition to the effects mentioned above, chemotherapeutic agents can also drive a myCAF phenotype in stromal fibroblasts. Exposure to these drugs can increase the amount of secreted ECM by CAFs that, among other effects, can form a physical barrier and prevent the drug from reaching the cancer cells
[85][162][85,162]. Chemotherapy-induced secretion of exosomes by fibroblasts can also play an important role in resistance to chemotherapy. In PDAC, Richards et al. showed that fibroblast-derived exosomes regulate EMT pathways in cancer cells in a Snail-dependent way and that targeting this pathway increased the sensitivity of cancer cells to chemotherapy
[163]. Transfer of miRNAs from CAFs to cancer via secreted extracellular vesicles has also been shown to be induced by chemotherapy and to promote resistance
[164].
Similar effects have been described for fibroblasts exposed to radiotherapy
[151]. Strong desmoplastic reactions often characterise the irradiated areas. This is a consequence of fibroblast activation and enhanced deposition of ECM
[165]. Indeed, an enrichment in αSMA-positive fibroblasts can be found in chemo and radiotherapy treated tumours
[158][166][158,166] and stromal signatures are often associated with a worse outcome regarding disease free survival
[166][167][168][169][166,167,168,169]. High amounts of ECM can then activate pro-survival pathways in cancer cells and impair the response to radiotherapy as well as to chemotherapy, in an integrin β1-dependent manner
[87][88][170][171][172][87,88,170,171,172]. As with cytotoxic agents, radiation can drive a senescent phenotype in stromal fibroblasts, which is characterised by the expression of, among others, TGF-β
[154]. The presence of elevated levels of TGF-β can then not only increase ECM production but also drive an immunosuppressive environment
[154]. Tommelein et al. also found that irradiated fibroblasts could promote the survival of cancer cells via insulin-like growth factor receptor-1 (IGF1R) signalling. This pathway activation was driven by a senescence-like phenotype of the fibroblasts that was characterized by the secretion of IGF1 after treatment
[173].
Despite their intended specificity, targeted therapies can also directly affect and modulate the phenotype of stromal fibroblasts. In colorectal cancer, the treatment of tumours with an EGFR inhibitor led to an increased secretion of EGF by stromal fibroblasts and conferred resistant of neighbouring cancer cells to the treatment via activation of MAPK signalling
[174]. Matrix remodelling by fibroblasts exposed to BRAF inhibitors can also impair the response of cancer cells to the treatment. In this system, resistance was driven by elevated activation of integrin β1-FAK-Src signalling in malignant cells because of high ECM-production by fibroblasts
[64].
2.3.2. Indirect Effects in CAFs
Fibroblasts can further be indirectly modulated by therapy, since it has been described that cancer cells acquire a specific secretory profile after exposure to drugs, named therapy-induced secretome
[175]. In the same way that cytotoxic agents can drive the senescence of fibroblasts, the cancer cells exposed to these agents can also undergo therapy-induced senescence and acquire the aforementioned senescence-associated secretory phenotype (SASP)
[175], which then modulates the phenotype of fibroblasts. We have recently shown that cancer cells exposed to high doses of chemotherapy upregulate the expression of IFNβ1, which acted in a paracrine manner to drive a pro-tumorigenic state of fibroblasts that then drove the recovery of cancer cells after treatment
[176]. This upregulation of IFNβ1 after treatment with cytotoxic agents goes in line with previous studies that have shown that high levels of damage in cancer cells after treatment results in the activation of the STING/IRF3 pathway and drives IFNβ1 expression
[177][178][179][177,178,179]. Moreover, therapies such as chemotherapy and radiation can strongly modulate the immune milieu of the TME and, consequently, affect the profile of stromal fibroblasts
[74]. Radiation can result in vascular damage, which triggers an inflammatory response and, consequently, promotes myofibroblast differentiation
[151]. This vascular damage can also lead to hypoxia and increase the production of HIF1α
[180][181][180,181]. In colorectal cancer, it was shown that HIFα and TGFβ cooperate to induce hedgehog transcription factor GLI2 expression in tumour-initiating cancer cells, which drives stemness and chemoresistance
[62]. The increased secretion of TGF-β by melanoma cells after exposure to Vemurafenib, a BRAF-inhibitor, was also able to drive fibroblast activation. Fibroblasts were then shown to produce increased levels of ECM, but also growth factors such as NRG-1 and HGF. Combined, these factors promoted the survival of cancer cells to treatment
[182]. Apicella et al. described a non-autonomous cancer cell mechanism in non-small cell lung cancer (NSCLC) that drove resistance to targeted therapies. Interestingly, they show that in vivo resistant cancer cells become re-sensitized to the therapy when treated in vitro in the absence of the TME. Mechanistically, they show that cancer cells treated with TKIs targeting MET and EGFR secrete higher levels of lactate, which then instructs CAFs to secrete HGF, resulting in the non-responsiveness of cancer cells to the treatment
[58].
The effects of therapies in fibroblasts are vast and complex. Despite the attempts to understand these mechanisms, further investigations are required. All the studies thus far have addressed these alterations in a bulk-fashion and detailed single-cell studies are still lacking. Moreover, it is necessary to unravel whether the mechanisms described above are tumour type- or context-specific, to develop strategies to identify which patients would benefit from their inhibition and, importantly, at what time-point during treatment. Understanding these complex dynamics is hard, but it would provide essential insights for the targeting of this cell type for cancer treatment.
2.4. Targeting Fibroblasts in Cancer Treatment
Several strategies have been developed for the targeting of fibroblasts in cancer treatment. In pre-clinical models, attempts to (1) directly target fibroblasts, (2) target secreted molecules such as ECM or soluble signalling molecules or (3) inhibit pro-tumorigenic signalling pathways have been described. Regardless of the strategy used the major goals have been to either eliminate stromal fibroblasts in general or certain subpopulations from the tumour, or to normalise the stroma/tumour-stroma interactions to ensure that the crosstalk between fibroblasts and cancer cells or the immune milieu does not support tumour progression, invasion or therapeutic failure. A summary of these strategies is described in
Figure 2.
Figure 2. Strategies for cancer therapy targeting fibroblasts and their interactions in the TME. Several approaches have been proposed to target CAFs, including direct targeting and elimination of specific subpopulations of fibroblasts, namely FAP-positive cells. Therapies against secreted factors and their respective signalling, such as ECM or cytokine signalling inhibitors have also been developed to improve immunity and to block pro-tumorigenic interactions between CAFs and their surroundings.
2.4.1. Direct Targeting of Fibroblasts
Numerous attempts of targeting fibroblasts have focused on their direct targeting via specific cell surface markers. Some early studies in PDAC have targeted αSMA-expressing fibroblasts. However, contrary to what had been expected, tumours in which this stromal population was eliminated grew at even higher rates, demonstrating that, at least in this tumour type, αSMA-positive fibroblasts restricted tumour growth
[28][29][28,29]. Similar attempts to eliminate or directly target fibroblasts in the TME have instead focused on the FAP expressing fibroblast population. In this case, regression of the tumour was indeed observed
[123][129][123,129]. Diverse FAP-targeting strategies have since been developed and tested in pre-clinical models, including genetic deletion
[123][129][123,129], molecular inhibitors that block FAP enzymatic activity
[129][183][184][185][186][129,183,184,185,186], anti-FAP monoclonal antibodies
[187] and FAP-antigen vaccination
[188], all with promising tumour-restraining results. Most of these studies correlate the observed effects with the impact of the treatment in the immune milieu. The tumours in which FAP was targeted were characterised by a reversion of the immunosuppressive environment and an increase in T-cell infiltration
[123][185][186][123,185,186]. Consequently, clinical trials targeting this protein have been undertaken to investigate the potential of such treatments in patients. The usage of blocking antibodies, namely sibrotuzumab (a humanised anti-FAP antibody, F19), proved to be safe in a phase I trial
[189][190][189,190], but failed to significantly improve overall survival. Similar results were obtained with the FAP-inhibitor PT100 (talabostat)
[191][192][193][194][191,192,193,194]. Even though the application of anti-FAP antibodies showed very limited clinical efficacy, these agents exhibited very good stroma-targeting properties
[190]. This has led to the development of antibody conjugates, in which FAP is used for the localised delivery of the conjugate. One such fusion antibody that is currently in a phase I clinical trial (NCT02627274) is called RO6874281, in which FAP is cross-linked with IL-2. The main goal of using this antibody is to activate T-cells in the TME
[195]. Other strategies involve the conjugation of FAP with other immune modulators, such as IL15
[196], co-stimulatory ligands as B7.2
[197] and CD40
[198], or immunotoxins
[199][200][199,200]. Moreover, FAP antibodies can be conjugated with agents that will directly induce apoptosis, such as the cytotoxic drug DM1, which have shown potent inhibitory activity in pre-clinical models
[187]. Finally, the ablation of FAP-expressing cells in the TME was also achieved by using CAR-T cells directed against FAP
[201][202][201,202].
In summary, a broad variety of strategies targeting FAP are available, although clinical trial results have been disappointing thus far. This might be improved with optimised strategies to directly target and eliminate FAP-positive cells, such as conjugated antibodies, vaccination or bispecific CAR-T cells. The enhanced immune response that has been observed in the pre-clinical models suggests that a combination of these strategies with other therapies, such as immune checkpoint inhibitors or chemotherapy, will likely be advantageous. Moreover, several studies clearly showed that these approaches are highly effective to target specific conjugates to the tumour site, which can be further explored.
Another cell surface marker that has been explored in pre-clinical models is GPR77. This protein, in combination with CD10, has been shown to identify a subpopulation of fibroblasts that is responsible for driving chemoresistance. The blockage of GPR77 using a neutralising antibody was shown to reduce the CSCs in tumours and enhance the response to chemotherapy
[60]. Moreover, since these fibroblasts were identified as being present in the tumour prior to treatment, the concomitant targeting of these with the chemotherapeutic regiments could likely provide a benefit for the patient. Further studies along these lines are required.
2.4.2. Targeting of the ECM
Stromal rich tumours can have impaired drug delivery as a result of the physical barrier presented by the ECM. Moreover, the ECM can activate pro-survival signalling pathways in tumour cells. This makes approaches targeting the ECM and/or its downstream signalling attractive for the treatment of a variety of tumours. Numerous pre-clinical studies blocking integrin signalling have shown that this axis is critical for the development of drug resistance and that its abrogation in combination with cytotoxic agents could improve therapy response and overall survival
[87][203][87,203]. A blocking antibody, FG-3019, that interferes with integrin signalling activation is being tested in a phase I/II clinical trial (NCT02210559). Moreover, the biosafety of defactinib, a small molecule inhibitor that targets FAK and thereby prevents downstream pathway activation in cancer cells, has been investigated in a phase I clinical trial (NCT02546531)
[204]. Direct targeting of ECM proteins, namely fibronectin
[205], tenascin C
[206] and hyaluronan
[138][207][208][138,207,208] have also been studied in pre-clinical models with promising results. The targeting of hyaluronan for degradation using a PEGylated enzyme, PEGPH20, in combination with chemotherapy showed positive results in early phase clinical trial in patients with PDAC
[209][210][209,210]. Losartan, a small molecule inhibitor that targets the angiotensin receptor and, consequently, leads to a decrease in hyaluronan levels, is currently tested in a phase I clinical trial (NCT03563248)
[211]. Another strategy employed in pre-clinical trials exploits the inhibition of matrix metalloproteinases (MMPs). These enzymes are critical players in the remodelling of the ECM
[212]. Unfortunately, disappointing results have emerged from all the MMP inhibitors tested in clinical trials thus far
[213]. Novel strategies are also emerging, with the use of CAR-T cells engineered to express heparanase, an enzyme that degrades ECM proteins, having shown promising pre-clinical results by enhancing T cell infiltration and anti-tumour activity
[214].
2.4.3. Targeting of Cytokines and Growth Factors
One of the most extensively studied and described pro-tumorigenic axes involved in the communication between cancer cells and their microenvironment is the IL-6/IL-6R/JAK-STAT3 pathway. Not surprisingly, attempts to target this pathway soon emerged and were tested in pre-clinical models
[215]. The targeting of this axis is threefold, since (1) it has been described to be involved in the activation of fibroblasts
[216][217][216,217], (2) its effect in tumour cells drives pro-tumorigenic states by, among others, modulating stemness and invasiveness
[218] and (3) it negatively regulates tumour-infiltrating immune cells
[219]. An IL-6 monoclonal antibody, siltuximab, was tested in a phase II clinical trial for the treatment of prostate cancer and demonstrated a safe profile but failed to improve patient outcome
[220]. The efficacy of this antibody is currently evaluated in a phase Ib/II trial for the treatment of advanced pancreatic cancer (NCT04191421). Furthermore, several clinical trials investigating the activity of molecules that inhibit downstream signalling of IL6-, namely ROCK and STAT3 inhibitors, have been undertaken. A dual ROCK-AKT inhibitor, AT13148, was tested in phase I, but failed to show a safe profile
[221]. The STAT3 inhibitor, AZD9150, was well tolerated in a phase I clinical trial
[222] and an investigation of its efficacy is undergoing.
Another major pro-tumorigenic molecule that is secreted by CAFs is CXCL12. This chemokine is mostly involved in the modulation of immune cells in the TME, and its targeting can alleviate immunosuppression and drive effective anti-tumour immunity. Therefore, the inhibition of this axis using antagonists or antibodies targeting its receptor, CXCR4, has been extensively studied
[223][224][223,224]. The efficacy of some of these molecules, namely the CXCR4 antagonist AMD3100, is currently evaluated in clinical trials for the treatment of several tumour entities, including haematological malignancies and advanced pancreatic cancer (
https://www.cancer.gov/about-cancer/treatment/clinical-trials/intervention/plerixafor, accessed on 1 June 2021), after having displayed a safe profile in phase I clinical trials
[225][226][225,226].
The inhibition of Shh and TGFβ has also been extensively studied in an attempt to ‘normalise’ the tumour stroma by reverting the activated state of fibroblasts. Moreover, these molecules have broad activities and are capable of regulating several other processes, as described in the previous sections. Despite the exciting results of Shh inhibition in pre-clinical studies
[52][61][227][52,61,227], evaluation of the FDA-approved inhibitors, saridegib and vismodegib, in early phase clinical trials showed very disappointing results with no improvement in disease-free survival (DFS) and overall survival (OS) (NCT01064622; NCT01088815)
[228]. To date, several TGFβ inhibitors have been developed and their efficacy studied using pre-clinical models
[229][230][231][229,230,231]. Following these results, clinical trials have been established and, while some have failed to demonstrate a significant overall benefit, other trials showed a benefit of inhibiting TGF-β signalling
[231].
Despite the many attempts to target the TME and to significantly improve patient outcomes, most of these strategies have fallen short. Improved combinatorial strategies will likely be required to achieve an effective inhibition of tumour growth. Moreover, most of the clinical trials enrol patients with heavily pre-treated tumours and advanced/metastatic disease, which can hinder the effectiveness of these drugs. The rational targeting of these axes in early treatment could provide an advantage by preventing the development of resistance and inhibiting the invasive properties in cancer cells. However, more studies are required to evaluate this hypothesis. Furthermore, increasing knowledge in CAF biology and the unravelling of subpopulation-specific markers will likely drive the development of more effective treatments as this might allow the targeting of particularly relevant subpopulations with enhanced specificity. Finally, the identification of biomarkers and the better stratification of patients will be necessary to see a potential improvement in disease-free survival and overall survival in particular patient groups, as with other targeted therapies in cancer therapy.