Open access peer-reviewed chapter

Perspective Chapter: Critical Role of Hedgehog in Tumor Microenvironment

Written By

Xing-Guo Li and Jer-Yen Yang

Submitted: 01 September 2022 Reviewed: 31 October 2022 Published: 24 January 2023

DOI: 10.5772/intechopen.108831

From the Edited Volume

Tumor Microenvironment - New Insights

Edited by Ahmed Lasfar

Chapter metrics overview

85 Chapter Downloads

View Full Metrics

Abstract

Hedgehog (Hh) signaling is a highly conserved pathway that plays a pivotal role during embryonic development. Mounting evidence has implicated Hh signaling in various types of cancer. Accordingly, inhibition of aberrant Hh signaling continues to be pursed across multiple cancer types -with some success in certain malignancies. In addition, with the renaissance of antitumor immunotherapy, an in-depth understanding of the molecular mechanisms underlying how the multifaceted functions of Hh signaling shape immunologically suppressive tumor microenvironment might be the key to unlocking a new era of oncological treatments associated with a reduced propensity for the development of drug resistance. Here, we focus on the latest advances regarding the immunological effects of misregulation of Hh signaling on tumor immunity. We also review the current status of clinically approved Hh inhibitors and dissect the mechanisms of drug resistance. Finally, we discuss the potential clinical applications that harness the immunomodulatory effects of Hh signaling not only to circumvent drug resistance, but also to achieve durable efficacy following immunotherapies, thus ultimately resulting in improved patient outcomes.

Keywords

  • hedgehog signaling
  • tumor microenvironment
  • immune cell
  • smoothened inhibitors
  • therapeutic targeting

1. Introduction

The Hedgehog (Hh) signaling pathway was discovered as a key regulator of organ development in Drosophila melanogaster by Christiane Nüsslein-Vollhard and Eric Wieschaus in the 1980s [1]. It was named after the gene locus associated with a spiky appearance of “hedgehog” phenotype in mutant Drosophila larve, findings based on which both investigators were awarded the Nobel Prize in Physiology or Medicine in 1995 “for their discoveries concerning the genetic control of early embryonic development,” together with Edward B. Lewis [2]. Since then, the Hh signaling has been extensively studied as a highly conserved evolutionary pathway to orchestrate embryonic development, cell growth and differentiation, homeostasis [3]. Unlike other classical signaling cascades, Hh signaling is almost silent in the adult organisms but reactivated in a few tissues such as the skin, during tissue regeneration and wound healing [3]. Not surprisingly, aberrant activation of this pathway has been demonstrated as a potent oncogenic driver to promote numerous hallmarks of cancer [4]. Therefore, the multifaceted role of Hh signaling may allow exploitation of this key pathway for novel and more effective cancer therapy [5].

Activation of Hh signaling is dependent on the primary cilium, a highly specialized organelle found on most vertebrate cells. Three Hh ligands, sonic hedgehog (Shh), desert hedgehog (Dhh), and Indian hedgehog (Ihh), are known to actuate the Hh pathway during embryonic and tissue development [6]. Whereas the expression patterns for Dhh and Ihh are tissue-specific, Shh has a broader expression pattern in various compartments and in multiple developmental stages [6]. In general, the Hh signaling is activated through either canonical or non-canonical mechanisms. In the canonical pathway, Hh ligands bind to the surface receptor Patched 1 (PTCH1), which alleviates the inhibitory effect of PTCH1 on a G-protein-coupled receptor (GPCR)-like protein, Smoothened (SMO), leading to migration of SMO to the tip of the cilium, which in turn signals suppressor of fused (SUFU) to release glioma-associated oncogene homolog proteins (GLIs). Finally, GLIs translocate into the nucleus, resulting in a signaling cascade through transcriptional regulation of Hh target genes [6]. Alternatively, GLI transcription factors can be activated through non-canonical mechanisms, which can be independently of PTCH1, SMO, or both [6]. Of note, mounting evidence has demonstrated that the signaling pathways that can induce non-canonical Hh signaling have been of known significance in oncogenesis, providing the mechanistic basis of the cross talk between Hh signaling and other signaling pathways to promote tumorigenesis, as well as the rationale for development of potential combination therapeutics [7, 8, 9, 10].

The discovery of PTCH mutations in basal cell nevus syndrome (BCNS, or Gorlin syndrome, or nevoid basal cell carcinoma [BCC] syndrome), a hereditary form of BCC, provides the first link between the Hh signaling and tumorigenesis [11, 12]. Other than BCC, emerging evidence has involved abnormal activation of Hh signaling in a variety of cancer types, such as medulloblastoma, breast cancer, pancreatic cancer, and lung cancer [13].

So far, three models have been proposed to elucidate the role of Hh signaling in oncogenesis where Hh signaling is over-activated through ligand production, autocrine, juxtacrine, or paracrine reception of the ligand, as well as cross talk between Hh signaling and complex intracellular signaling cascades [13]. First, in BCC and medulloblastoma, activating mutations of Hh pathway have been identified, such as inactivating mutations in PTCH or SUFU, and activating mutations in SMO, as shown in 85% of sporadic BCC or 30% of medulloblastoma, respectively [11, 12, 13, 14, 15, 16]. In this scenario, the autonomous activation of Hh signaling is independent of Hh ligands.

Second, Hh signaling is aberrantly activated through autocrine or juxtacrine ligand-dependent manner, where Hh is secreted and responded by the same or adjacent cells [13]. This category of cancers includes breast cancer, pancreatic cancer, lung cancer, prostate cancer, colorectal cancer, stomach and esophageal cancer, ovarian and endometrial cancer, melanomas, and gliomas [13]. Finally, in pancreatic cancer, prostate cancer, and colon cancer, Hh signaling is activated through a paracrine-dependent manner, where Hh ligands are secreted by tumor cells, whereas the PTCH receptor is expressed on stromal cells in the tumor microenvironment (TME). In this last model of Hh signaling activation, a feedback loop is generated, which allows the transmit of the growth-promoting signals from tumor cells to stromal cells and then back to tumor cells, leading to sustained tumor progression [17].

In the following sections, we will first highlight the key cellular components of TME involved in oncogenic Hh signaling to promote tumor progression. We will then review the current status of the FDA-approved and non-approved inhibitors of Hh signaling, as well as the molecular mechanisms of drug resistance. Finally, we will provide a critical evaluation of recent studies on the treatments combining immunotherapeutic strategies with approved Hh inhibitors and will propose potential strategies that could be applied to harness existing knowledge to overcome the drug resistance.

Advertisement

2. The role of Hh signaling in the TME

Emerging evidence has suggested that TME is not just a silent bystander, but rather an active player of tumor progression [18, 19]. The composition of TME not only varies between tumor types, but also is continuously evolving in the different stages of tumorigenesis. Hh signaling has been intensively studied with respect to the classical hallmarks of cancer [3, 4, 5, 6]. In contrast, its role in the modulation of TME has only become evident in recent studies [20, 21, 22].

2.1 Immune cells

The adaptive and innate immune systems cooperate to form a highly proficient immune surveillance machinery that can identify and eradicate genetically altered cells to prevent tumorigenesis. Tumor-infiltrating leukocytes (TILs), including T and B lymphocytes, monocytes and macrophages, myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), and natural killer (NK) cells, play diverse roles in tumor progression through interactions and production of cytokines, chemokines, and growth factors to support or suppress tumor growth and metastasis [20, 21]. There is increasing evidence from multiple experimental models that demonstrate an important and multifaceted role of Hh signaling in the modulation of immune cell functions. Aberrant Hh signaling induces a hostile, immunosuppressive microenvironment to dampen an effective antitumor immune response.

Regulatory T cells (Tregs) control the activity of effector immune cells such as granzyme B-expressing CD8+ T cells and NK cells by secreting anti-inflammatory cytokines such as TGF-β and interleukin-10 (IL-10) [23]. The immune modulatory role of Hh signaling in T cells is evidenced by recent studies demonstrating that Hh signaling may directly regulate the expression and activity of TGF-β. Treg infiltration has been described for Hh-associated tumors, such as BCC [24], and medulloblastoma [25, 26, 27, 28].

Intriguingly, elevated Treg infiltration is accompanied by an increase of TGF-β within intra- and peri-tumoral skin in a human UV-exposed facial BCC model [29]. In line with the putative immunosuppressive phenotype of Hh signaling, genetic abrogation of T-cell TGF-β signaling mitigated tumor progression in a transgenic medulloblastoma mouse model overexpressing smoothened A1 (SmoA1), an obligatory and conserved Hh signal transducer [25]. In this study, TGF-β signaling blockade led to nearly abolishment of Tregs and licensing of CD8 cytotoxic T lymphocytes for antitumor immunity [23].

Mechanistically, GLI2, an Hh effector, has been shown to directly activate the expression of TGF-β in human Tregs [30]. Thus, it has been proposed that Hh signaling may help generate a feed-forward loop where TGF-β induces the inversion of CD4+ T cells to Tregs, which in turn secrete high levels of TGF-β, leading to enforcement of the continued presence of immunosuppressive Tregs in the tumor microenvironment [31].

Myeloid cell infiltration has been described in multiple cancer entities where tumors may benefit from myeloid cells-mediated immunosuppression. The role of Hh signaling in the tumor-promoting function of myeloid cells has been postulated based on observations in multiple models of Hh-induced tumors. First, in a murine SMO-induced BCC model, tumor growth appears to be enhanced by the recruitment of immunosuppressive myeloid derived suppressor cells (MDSCs), accompanied by a reduction of effector lymphocytes in the tumor lesions [32]. This is mediated by the TGF-β-CCL2 (C-C motif chemokine ligand 2) axis secreted by oncogenic SMO- expressing keratinocytes and the CCL2 receptor expressed by MDSCs. In vivo, pharmacological suppression of the CCL2 receptor expression decreased infiltration of MDSCs and resulted in reduced tumor growth, indicating an immunosuppressive phenotype by the oncogenic Hh signaling [33]. Likewise, there is also strong evidence for immunosuppressive function of myeloid cells in Hh-associated medulloblastomas, which are characterized by high myeloid infiltration. For example, gene expression profiling of human Hh medulloblastoma tumors showed enrichment for an M2-like gene expression profile, consistent with immunosuppressive functions of myeloid cells [34]. Moreover, an inverse correlation has been observed between expression of M2-like markers (such as CD163) and survival of human Hh medulloblastoma patients [34].

Along these lines, the notion of an immunosuppressive function of Hh signaling was further affirmed by two recent studies in Hh-induced medulloblastomas. In a mouse model of Hh medulloblastoma (Ptch1+/−; Tp53−/−), Dang et al. showed decreased T-cell proliferation in a co-culture system of tumor-infiltrating myeloid cells and ex vivo stimulated T cells [35]. Mechanistically, the immunosuppressive phenotype appears to be mediated by CCL2. Genetic knockout of CCL2 receptor not only decreased infiltration of monocyte-derived macrophages but also increased levels of CD8+ T cells in tumors [35]. Likewise, in another mouse model of Hh-induced medulloblastoma (Atoh1-SmoM2), pharmacological inhibition of colony stimulating factor 1 receptor (CSF1R) depleted macrophages and microglia, resulting in delayed tumor growth and prolonged mouse survival [36]. These recent studies support the notion of a tumor- promoting function of macrophages, which are consistent with an early study in another Hh-associated medulloblastoma tumor model, where the presence of MDSCs increases infiltration of Tregs and reduces the number of effector T cells [37]. Interestingly, infiltrating myeloid cells have been described as the predominant source of PD-L1 expression in a mouse model of Hh-induced medulloblastoma where the binding of PD- L1 to PD-1 on effector T cells resulted in T-cell exhaustion and immune escape of tumor cells [38]. Furthermore, an analysis of an immunocompetent breast cancer xenograft mouse model showed that inhibition of Hh signaling (SMO inhibitor vismodegib) led to reduced infiltration of immunosuppressive myeloid cells in the tumors, accompanied by an increase of effector CD8+ T cells and M1 macrophages [39].

2.2 Tumor-associated astrocytes (TAAs)

Astrocytes, the most abundant type of glial cells in the brain, are integral partners with neurons in the regulation of neuronal development and brain function. Hh signaling has emerged as a critical player to support astrocyte-mediated modulation of neuronal activity [40, 41, 42]. A recent series of elegant work supports a key role of tumor-associated astrocytes (TAAs) in promoting tumor growth and metastasis through distinct signaling, including Hh pathway [43, 44, 45, 46]. First, TAAs were shown to secret the ligand Shh, which is required for maintaining cell proliferation of Hh-activated medulloblastoma through a Smo-dependent, but Gli1-independent manner, despite the absence of its primary receptor Ptch1. Of note, ablation of TAAs blocked tumor growth [43]. Furthermore, a recent study at single-cell resolution demonstrated that Hh-induced medulloblastoma cells can transdifferentiate into interleukin-4 (IL-4)-secreting TAAs, which in turn stimulates tumor-associated microglia to release insulin-like growth factor 1 (IGF1) to promote tumor progression [44]. Similarly, medulloblastoma-associated astrocytes have recently been shown to produce high levels of CCL2, a tumor-promoting cytokine shown to drive stemness maintenance and proliferation of disseminated tumor cells [45] and to promote metastasis [47]. Moreover, using single-cell RNA sequencing and lineage tracing analyses, Guo et al. investigated cellular origin of TAAs in a mouse model for relapsed Hh-activated medulloblastoma driven by Ptch1 knockout [46]. This study has elegantly demonstrated that TAAs are predominantly derived from the transdifferentiation of tumor cells in relapsed MB, but not in primary MB, thus establishing the distinct cellular sources of astrocytes [46]. Interestingly, this study revealed that such transdifferentiation of medulloblastoma cells to TAAs depends on bone morphogenetic proteins (BMPs) and that pharmacological inhibition of BMP signaling repressed transdifferentiation and suppressed tumor relapse [46]. It remains to be determined what drives these transdifferentiation events and what intrinsic and extrinsic mechanisms, beyond Hh and BMP signaling, regulate the potential cooperation of TAAs and microglia in promoting the immunosuppressed state of medulloblastoma.

2.3 Cancer-associated fibroblasts (CAFs)

Cancer-associated fibroblasts (CAFs), the most abundant stromal cells in TME, have emerged as a central player in cancer progression and metastasis [48]. Through diverse phenotypes, origins, and functions, CAFs modulate the cross talk between inflammation and tumorigenesis and contribute to therapeutic resistance by producing various cytokines, chemokines, growth factors, and matrix-degrading enzymes [49].

There is increasing evidence indicating that CAF populations that support or suppress tumor growth and progression through stroma-specific Hh activation have been detected in multiple tumor types, including pancreatic cancer, colon cancer, and bladder cancer [50]. Recent advances in single-cell technologies have enabled detailed characterization of the heterogeneity and plasticity of differential CAF subsets, supporting a new therapeutic strategy in which tumor-supporting CAFs are reprogrammed into tumor-suppressing CAFs [50]. In pancreatic ductal adenocarcinoma (PDAC), Hh signaling pathway is activated in CAFs via a paracrine mechanism and has been associated with pancreatic tumorigenesis [49]. Initial studies indicated that inhibition of Hh pathway impaired tumor growth and sensitized tumors to chemotherapy in multiple PDAC models [51, 52, 53, 54, 55, 56]. However, recent studies have challenged the concept of tumor-promoting CAFs. In the context of an oncogenic Kras-driven mouse PDAC model, conditional deletion of Shh, the predominant Hh ligand expressed in pancreas, led to cachexia and to poorly differentiated and highly vascularized tumors [57].

Moreover, by using three distinctly genetically engineered mouse PDAC models, another study showed that pharmacologic inhibition of Hh pathway activity accelerated rather than delayed progression of oncogenic Kras-driven disease by affecting the balance between epithelial and stroma elements, leading to suppression of stromal desmoplasia but accelerated growth of pancreatic intraepithelial neoplasia [58]. These contradictory findings indicate that Hh signaling may play pleiotropic roles in PDAC progression. Interestingly, by using a combination of pharmacologic inhibition, gain- and loss-of-function genetic experiments, cytometry by time-of-flight, and single-cell RNA sequencing, a more recent study defines dosage-dependent effects of Hh signaling on the composition and function of CAFs in PDAC microenvironment [59]. Hh signaling is uniquely activated and differentially elevated in CAFs, with higher levels in myofibroblastic CAFs (myCAF) compared with inflammatory CAFs (iCAF) in both mouse and human PDAC. Driving high levels of Hh signaling promotes tumor growth, whereas Hh pathway inhibition alters the ratio of myCAF/iCAF populations, which is accompanied by a decrease in cytotoxic T cells and an expansion in regulatory T cells, thus altering the composition of CAFs, and shifting the inflammatory response toward a more immunosuppressive phenotype [59]. Given the differential functional implications for CAF subpopulations, changes in the ratio of CAF subtypes may lead to distinct antitumor outcomes. Consistent with, recent studies demonstrated a possible negative impact of current Hh pathway inhibitors on antitumor response in clinical trials, which were largely unsuccessful or even detrimental to patient health [60, 61]. Further understanding of the roles of Hh signaling in CAFs may open the possibility for more effective combination cancer therapies.

Advertisement

3. Therapeutic targeting Hh signaling in cancers

Given the multifaceted role of Hh signaling in cancer, inhibitors of Hh pathways have emerged as an important class of anticancer agents. These compounds fall into three main categories: Hh ligand inhibitors, SMO inhibitors, and GLI inhibitors [62]. Despite extensive efforts devoted to the discovery of Hh signaling inhibitors, so far only three drugs have been approved by the Food and Drug Administration (FDA), all targeting the upstream receptor of Hh signaling SMO, a membrane protein of the GPCR protein family [62].

3.1 FDA-approved inhibitors

To date, three SMO inhibitors, vismodegib, sonidegib, and glasdegib, have been FDA approved in 2012, 2015, and 2018, respectively, for cancer treatment. Cyclopamine, the first SMO antagonist, is a naturally occurring alkaloid found in the corn lily [63] later proved to bind to SMO and to inhibit activation of downstream Hh target genes [64].

Extensive efforts have been made to develop alkaloid derivatives to increase the bioavailability, sensitivity, and specificity of cyclopamine to target SMO [65]. Vismodegib (GDC-0449 or Erivedge), the first cyclopamine derivative and Hh pathway–targeting drug, is currently approved for the treatment of patients with locally advanced or metastatic BCC (US FDA). Compared to cyclopamine, vismodegib shows a higher potency and more favorable pharmacological properties [62]. The approval of vismodegib was based on results from the pivotal phase II ERIVANCE trial (ClinicalTrials.gov, NCT00833417) showing that vismodegib substantially shrank tumors or healed visible lesions (objective response rate, ORR) in 43% of patients with locally advanced BCC and 30% of patients with metastatic BCC, at 21 months, with a median progression-free survival (PFS) duration of 9.5 months for both metastatic and locally advanced BCC patients [66, 67]. Up to the completion of this manuscript, there have been 86 clinical trials for vismodegib, both monotherapy and combination, in various cancer types (ClinicalTrials.gov).

Sonidegib (Erismodegib, NVP-LDE-225, LDE-225, Odomzo) is another cyclopamine-derived SMO antagonist discovered in 2010 through an in vitro, high-throughput screen, showing high tissue penetration and bioavailability, as well as the ability to cross the blood-brain barrier [68]. In 2015, sonidegib became the second SMO inhibitor approved for patients with locally advanced or recurrent BCC (US FDA). The approval of sonidegib was based on results from a multicenter, randomized, double-blind phase II BOLT trial (ClinicalTrials.gov, number NCT01327053), which showed the objective response rates of 38% and 43% in the 800 and 200 mg dosage groups, respectively after 30 months in patients with locally advanced BCC and the objective response rates of 17% and 15%, respectively in those with metastatic BCC [69]. Up to August 2022, there are 46 clinical trials for sonidegib in cancer treatment (ClinicalTrials.gov).

A third FDA-approved inhibitor of Hh signaling is glasdegib (PF-04449913, Daurismo), a benzamide derivative discovered in 2012 with high potency and oral bioavailability [70]. In 2018, glasdegib was approved for combination treatment with low-dose cytarabine arabinoside (LDAC) for patients with acute myeloid leukemia unsuitable for intensive chemotherapy. The approval of glasdegib was based on the results of the phase II BRIGHT 1003 trial (ClinicalTrials.gov, NCT01546038) showing the median overall survival of 8.8 months with glasdegib/LDAC as compared to 4.9 months with LDAC. Furthermore, 17.0% and 2.3% of patients in the glasdegib/LDAC and LDAC arms, respectively, achieved complete remission [71]. Up to this point, there have been 26 clinical trials for glasdegib in various cancer types (ClinicalTrials.gov).

3.2 Resistance mechanisms to FDA-approved inhibitors

The first retrospective study on drug resistance to SMO inhibitor therapy was reported in 2012 where 21% of BCC patients treated with vismodegib developed drug resistance, with a mean tumor recurrence time of 56.4 weeks in clinical examination [72]. Ever since, resistance to SMO antagonists has been observed in patients who never respond to SMO inhibitor therapy (primary resistance), as well as in those who initially respond but later develop resistance to SMO inhibitors (acquired resistance) [73]. Mechanistically, a number of models have been proposed to explain the basis of drug resistance to SMO inhibitor therapy. First, genetic analysis of resistant tumors has revealed mutations of SMO, loss of SUFU, and amplification of GLIs or Hh target genes, such as CCND1 and GLI1 [5, 10]. Second, accumulating evidence supports the notion that the resistance can be driven through the non-canonical Hh signaling, accompanied by the concurrent activation of other oncogenic signaling pathways, such as AP-1 and TGF-β signaling [74], RhoA signaling [75], and RAS-MAPK signaling [76]. Finally, a new mechanism has recently been uncovered to contribute to drug resistance through loss of primary cilia [77, 78]. This was supported by both preclinical and clinical evidence. In Hh-dependent medulloblastoma, recurrent mutations in oral facial digital syndrome 1 (OFD1), a culprit gene led to loss of cilia, and thereby caused resistance to SMO inhibitors [78]. Importantly, sequencing data analysis from resistant BCC patients showed recurrent mutations in ciliary genes, providing clinical relevance of this new mechanism [77]. Therefore, a better understanding of cilia-regulating signaling pathways in resistant cancer may open up a new route to reintroduce cilia to sensitize resistant cancer cells to SMO inhibitors. Taken together, several strategies have been proposed to overcome the drug resistance through targeting the underlying mechanisms. These approaches include: (1) develop second-generation SMO inhibitors to retain anticancer activities that are not affected by the resistance-inducing mutations [5]; (2) target downstream components of SMO, such as GLIs (see below, non- approved inhibitors), or signaling molecules involved in the non-canonical Hh signaling pathway [8].

3.3 Non-FDA-approved inhibitors

Multiple novel inhibitors targeting SMO have been shown to be effective in preclinical models [5] and are now in active clinical trials, either monotherapy or combination for various cancer types. These compounds include saridegib (patidegib, IPI-926), taladegib (LY2940680), and BMS-833923 (XL139) (ClinicalTrials.gov). On the other hand, even though GLI1 antagonists are not as extensive as those targeting SMO, mounting evidence has shown that targeting the Hh signaling at the level of its final effector, GLI1, is a promising strategy to overcome resistance to currently available SMO inhibitors [79, 80]. In this regard, the promising pharmacological potential of direct and indirect GLI inhibitors, as well as GLI antagonists derived from natural products, has been in active investigation at the preclinical or clinical phase. It is anticipated that future study on these compounds will help develop new strategies tackling resistant mechanisms and tumor heterogeneity [81].

Advertisement

4. Hh signaling and antitumor immune response

In 2018, James P Allison and Tasuku Honjo were awarded the Nobel Prize in Physiology or Medicine “for their discovery of cancer therapy by inhibition of negative immune regulation” [82]. Although this breakthrough in cancer immunotherapy has revolutionized cancer treatment, only a subset of patients elicit favorable responses and most immunologically cold solid tumors are not responsive [83]. Given the immunosuppressive function of Hh signaling, inhibitors of Hh signaling pathway may hold promise in converting nonresponsive cold tumors into responsive hot ones, which may subsequently allow nonresponders to benefit from immunotherapies. Notably, clinically approved Hh inhibitors, as well as non-approved inhibitors, have been in active preclinical and clinical trials for combined therapies, including immunotherapies.

The first clinical trial with Hh inhibitors in combination with immune checkpoint inhibitors was conducted in 16 patients with advanced BCC (clincialtrial.gov, NCT02690948). This trial showed that pembrolizumab (PD-L1 inhibitor) is active against BCCs. Although the two groups of pembrolizumab with or without vismodegib were not directly compared, the response rate for the combination group was not superior to the monotherapy group [84]. Of note, most patients with advanced BCC progress on or are intolerant to Hh inhibitor therapy despite objective response rates of 30–60% [66, 67, 68, 69, 85]. Until Feb 9, 2021, when cemiplimab, a PD-1 antibody, was approved by the US FDA fully for patients with locally advanced BCC, and accelerated for patients with metastatic BCC, after treatment with Hh inhibitors, or for whom Hh inhibitors are not appropriate [86], there was no standard second-line treatment option for these BCC patients [72]. A recent clinical trial study provides the first report to show clinically meaningful antitumor activity of cemiplimab in patients with BCC after Hh inhibitor therapy ([87], clinicaltrials.gov, NTC03132636). In this trial, the efficacy and safety of cemiplimab were evaluated in patients with locally advanced BCC or metastatic BCC who had previously been treated with an Hh inhibitor. Among the efficacy population (n = 121), centrally reviewed objective response was observed in 31% of patients with estimated duration of response exceeding 1 year in 85% of responders [87].

Importantly, this study also showed that the safety profile was consistent with what is known for immune checkpoint class of drugs, even considering the advanced age of the patient population in the present study [87]. These findings demonstrate the efficacy of immune checkpoint blockade in treating BCC in patients who had previously received Hh inhibitor therapy, thus opening a new horizon for treatment of the many patients who discontinue Hh inhibitor therapy due to disease progression, toxicity, or drug resistance. Moreover, a recent case report demonstrated an impressive response to cemiplimab in a sonidegib-resistant giant basosquamous carcinoma, one form of BCC [88]. Finally, a dozen of clinical trials have been initiated to investigate the combination treatment of anti-PD-1, PD-L1, and CTLA-4 monoclonal antibody therapy with first-line Hh inhibitors in patients with a variety of cancer types (see Table 1). The outcome of these trials will not only inform about whether combinatorial treatments can increase the efficacy and duration of antitumor response, but also provide insights into the optimal customized regimen to circumvent resistance to Hh inhibitors.

SMO inhibitorCombinationCancer TypeEnrollmentPhaseStatusNCT #
Vismodegib (GDC-0449 or Erivedge)+ VEGF-A antibody and chemotherapyMetastatic Colorectal Cancer199Phase 2CompletedNCT00636610
+ Anti-hormone therapyProstate Cancer10Phase 1|2TerminatedNCT01163084
+ ChemotherapyPancreatic Cancer118Phase 1|2CompletedNCT01064622
+ VEGF-A antibody and chemotherapyOvarian Cancer|Basal Cell
Carcinoma|Metastatic
Colorectal Cancer
19Phase 2CompletedNCT00959647
+ Notch inhibitorBreast Cancer13Phase 1TerminatedNCT01071564
+ ChemotherapyGastric Cancer124Phase 2CompletedNCT00982592
+ ChemotherapyPancreatic Cancer25Phase 2CompletedNCT01195415
+ ChemotherapyMyelodysplastic Syndromes,38Phase 2TerminatedNCT01880437
+ Notch inhibitorSarcoma78 Phase1|2CompletedNCT01154452
+ Photodynamic therapyBasal Cell Nevus Syndrome24Phase 2CompletedNCT01556009
+ IGF1R antibody and chemotherapySmall Cell Lung Carcinoma168Phase 2CompletedNCT00887159
+ ChemotherapyPancreatic Adenocarcinoma21Phase 1UnknownNCT01713218
+ ChemotherapyMedulloblastoma24Phase 1|2TerminatedNCT01601184
+ ChemotherapyMetastatic Pancreatic Cancer98Phase 2CompletedNCT01088815
+ DNMT inhibitorAcute Myeloid Leukemia40Phase 2UnknownNCT02073838
+ mTOR inhibitorPancreatic Cancer31Phase 1CompletedNCT01537107
+ PD1 blockadeSkin Basal Cell Carcinoma16Phase 1|2CompletedNCT02690948
+ ChemotherapyBreast Cancer40Phase 2UnknownNCT02694224
+ Radiation therapyBasal Cell Carcinoma24Phase 2CompletedNCT01835626
+ Radiation therapyCarcinoma, Basal Cell14Phase 2TerminatedNCT02956889
+ PD1/CTLA4 blockadeBasal Cell Nevus Syndrome0Phase 2WithdrawnNCT03767439
+ Tyrosine kinase inhibitorBasal Cell Carcinoma84Phase 2RecruitingNCT04416516
+ Tyrosine kinase inhibitors and PARP inhibitorsMiscellaneous950Phase 2RecruitingNCT02925234
+ PDL1 blockade and Tyrosine kinase inhibitorsMiscellaneous676Phase 2Active, not recruitingNCT02091141
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyLymphoma, Non-Hodgkin720Phase 2RecruitingNCT03297606
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyCancer of Unknown Primary Site790Phase 2RecruitingNCT03498521
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyMiscellaneous384Phase 2RecruitingNCT04591431
+ Targeted therapy and chemotherapyGlioblastoma, Adult350Phase 1|2RecruitingNCT03158389
+ Targeted therapy and chemotherapyMeningioma124Phase 2RecruitingNCT02523014
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyMiscellaneous300Phase 2RecruitingNCT04341181
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyMiscellaneous6452Phase 2RecruitingNCT02465060
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyMiscellaneous 40Phase 1RecruitingNCT03878524
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyMiscellaneous131Phase 2Not yet recruitingNCT05238831
+ PDL1 blockade, Tyrosine kinase inhibitors and chemotherapyAdvanced Cancer|Solid Tumor250Phase 2RecruitingNCT05159245
+ Radiation therapy and chemotherapyMedulloblastoma660Phase 2RecruitingNCT01878617
+ PDL1 blockadeCancer Metastatic1000Phase 2RecruitingNCT04817956
+ ChemotherapyPancreatic Cancer55Phase 1Active, not recruitingNCT00878163
Sonidegib (Erismodegib, NVP-LDE-225, LDE-225, Odomzo)+ Radiation therapy and chemotherapyMedulloblastoma205Phase 2Not yet recruiting NCT04402073
+ ChemotherapyLung Cancer19Phase 1CompletedNCT01579929
+ JAK inhibitorMiscellaneous50Phase 1|2CompletedNCT01787552
+ ChemotherapyPancreatic Ductal Adenocarcinoma23Phase 1|2TerminatedNCT01431794
+ ChemotherapyMyelodysplastic Syndrome63Phase 1CompletedNCT02129101
+ mTOR kinase inhibitorEsophageal Cancer25Phase 1CompletedNCT02138929
+ ChemotherapyPlasma Cell Myeloma28Phase 2CompletedNCT02086552
+ ChemotherapyPancreatic Cancer78Phase 1|2CompletedNCT02358161
+ Tyrosine kinase inhibitor and chemotherapyChronic Myelogenous Leukemia11Phase 1CompletedNCT01456676
+ Tyrosine kinase inhibitor and chemotherapyMiscellaneous108Phase 1RecruitingNCT03434262
+ Tyrosine kinase inhibitorMiscellaneous120Phase 1CompletedNCT01576666
+ ChemotherapyAdvanced Breast Cancer12Phase 1CompletedNCT02027376
+ Tyrosine kinase inhibitorCarcinoma, Basal Cell10Phase 2TerminatedNCT02303041
+ ChemotherapyPancreatic Cancer18Phase 1CompletedNCT01487785
+ PD1 blockadeMiscellaneous45Phase 1RecruitingNCT04007744
Neoadjuvant + SurgeryBasal Cell Carcinoma10Phase 2RecruitingNCT03534947
+ ChemotherapyMultiple Myeloma7Phase 2TerminatedNCT02254551
+ ChemotherapySolid Tumor|Ovarian Cancer30Phase 1CompletedNCT01954355
+ ChemotherapyPancreatic Cancer39Phase 1CompletedNCT01485744
+ ChemotherapyProstate Cancer0Phase 1WithdrawnNCT02182622
+ PD1 blockadeBasal Cell Carcinoma20Phase 2RecruitingNCT04679480
Glasdegib (PF-04449913, Daurismo)+ ChemotherapyGlioblastoma75Phase 1|2Active, not recruitingNCT03466450
+ ChemotherapyAcute Myelogenous Leukemia30Phase 2RecruitingNCT04231851
+ Antibody-drug conjugateAcute Myeloid Leukemia414Phase 3RecruitingNCT04168502
+ ChemotherapyACUTE MYELOID LEUKEMIA1Phase 2TerminatedNCT04051996
+ ChemotherapyAcute Myeloid Leukemia15Phase 3Active, not recruitingNCT04842604
+ ChemotherapyMyelodysplastic Syndrome73Phase 1CompletedNCT02367456
+ ChemotherapyLeukemia, Myeloid, Acute730Phase 3CompletedNCT03416179
+ ChemotherapyAcute Myeloid Leukemia0Phase 1WithdrawnNCT04655391
+ Antibody-drug conjugateAcute Myeloid Leukemia28Phase 3TerminatedNCT04093505
+ PD1 blockade, antibody-drug conjugate and chemotherapyAcute Myeloid Leukemia138Phase 1|2Active, not recruitingNCT03390296
+ ChemotherapyAcute Myeloid Leukemia48Phase 1Active, not recruitingNCT02038777
+ ChemotherapyLeukemia, Myeloid, Acute0WithdrawnNCT04230564
+ ChemotherapyAcute Myeloid Leukemia255Phase 2CompletedNCT01546038
+ ChemotherapyAdult Acute Myeloid Leukemia75Phase 2RecruitingNCT03226418
+ ChemotherapySoft Tissue Sarcoma960Phase 3RecruitingNCT03784014
+ Chemotherapy and radiation therapyGlioblastoma30Phase 1|2Not yet recruitingNCT03529448
Saridegib (patidegib, IPI-926)+ ChemotherapyMetastatic Pancreatic Cancer122Phase 1|2CompletedNCT01130142
+ ChemotherapyPancreatic Cancer15Phase 1CompletedNCT01383538
+ Tyrosine kinase inhibitorHead and Neck Cancer9Phase 1CompletedNCT01255800
Taladegib (LY2940680)+ Chemotherapy and radiation therapyEsophageal Adenocarcinoma7Phase 1|2CompletedNCT02530437
+ ChemotherapySmall Cell Lung Carcinoma26Phase 1|2TerminatedNCT01722292
+ Chemotherapy and CDK inhibitorsBreast Cancer|Colon Cancer|Cholangiocarcinoma|Soft Tissue Sarcoma94Phase 1CompletedNCT02784795
BMS-833923 (XL139)+ Tyrosine kinase inhibitorLeukemia33Phase 1|2CompletedNCT01218477
+ Tyrosine kinase inhibitorLeukemia70Phase 2TerminatedNCT01357655
+ ChemotherapySmall Cell Lung Carcinoma5Phase 1CompletedNCT00927875
+ ChemotherapyStomach
Neoplasms|Esophageal
Neoplasms 39
Phase 1CompletedNCT00909402
+ Proteasome inhibitorsAdvanced Cancer, Various, NOS27Phase 1CompletedNCT00884546

Table 1.

Combination therapy of SMO inhibitors under clinical trials.

Data from clinicaltrials.gov (accessed on 2022/8/22).

Comparatively a few recent studies have indicated possible negative effects of the current Hh inhibitor therapy on antitumor immunity [89]. For instance, blockade of SMO signaling may inhibit formation of the immunological synapse [90]. Administration of SMO inhibitors caused the functional disruption of the immunological synapse, leading to the loss of T-cell effector activity [90]. Even though it remains unclear whether Hh inhibitor therapy may impede cytotoxic T-cell killing in cancer patients, a pilot clinical trial study of vismodegib in combination with pembrolizumab did not suggest additive clinical activity [84]. In the clinical context, there is an emerging paradigm that immunotherapy may show the greatest activity when administered early in the natural history of cancers. Further studies are warranted to evaluate the efficacy and duration of immune checkpoint blockade before Hh inhibitor therapy.

Advertisement

5. Conclusions

The Hh signaling pathway has attracted extensive research attention as a key player to contribute to the progression of a variety of human cancer types. With an in-depth understanding of the molecular mechanisms underlying the role of Hh signaling in tumorigenesis, enormous efforts have been made to develop specific inhibitors targeting molecular components of this pathway. Consequently, cancer therapy has undergone a paradigm shift from eradicating tumor cells to multidimensional targeting and normalizing tumor cells and TME. Herein, we reviewed the multifaceted function of Hh signaling in shaping immunologically suppressive TME to promote tumor progression, provided an up-to-date status of active clinical trials of FDA approved Hh inhibitors, and finally, highlighted possible therapeutic interventions that harness the immunomodulatory effects of Hh signaling not only to overcome drug resistance, but also to achieve durable efficacy following immunotherapies.

Advertisement

Acknowledgments

This book chapter was in part of supported by MOST 110-2311-B-039-001, MOST 111-2320-B-039-050 and CMU109-YT-03 (X.-G.L.) and MOST 111-2320-B-039-015 and CMU109-YT-02 (J.-Y.Y.).

References

  1. 1. Nüsslein-Volhard C, Wieschaus E. Mutations affecting segment number and polarity in drosophila. Nature. 1980;287(5785):795-801. DOI: 10.1038/287795a0
  2. 2. The Nobel Prize in Physiology or Medicine. 1995. Available from: https://www.nobelprize.org/prizes/medicine/1995/nusslein-volhard/facts/
  3. 3. Briscoe J, Thérond PP. The mechanisms of hedgehog signalling and its roles in development and disease. Nature Reviews. Molecular Cell Biology. 2013;14(7):416-429. DOI: 10.1038/nrm3598
  4. 4. Wu F, Zhang Y, Sun B, McMahon AP, Wang Y. Hedgehog signaling: From basic biology to cancer therapy. Cell Chemical Biology. 2017;24(3):252-280. DOI: 10.1016/j.chembiol.2017.02.010
  5. 5. Nguyen NM, Cho J. Hedgehog pathway inhibitors as targeted cancer therapy and strategies to overcome drug resistance. International Journal of Molecular Sciences. 2022;23(3):1733. DOI: 10.3390/ijms23031733
  6. 6. Kong JH, Siebold C, Rohatgi R. Biochemical mechanisms of vertebrate hedgehog signaling. Development. 2019;146(10):dev166892. DOI: 10.1242/dev.166892
  7. 7. Brechbiel J, Miller-Moslin K, Adjei AA. Crosstalk between hedgehog and other signaling pathways as a basis for combination therapies in cancer. Cancer Treatment Reviews. 2014;40(6):750-759. DOI: 10.1016/j.ctrv.2014.02.003
  8. 8. Pietrobono S, Gagliardi S, Stecca B. Non-canonical hedgehog signaling pathway in cancer: Activation of GLI transcription factors beyond smoothened. Frontiers in Genetics. 2019;10:556. DOI: 10.3389/fgene.2019.00556
  9. 9. Li YH, Luo J, Mosley YY, Hedrick VE, Paul LN, Chang J, et al. AMP-activated protein kinase directly phosphorylates and destabilizes hedgehog pathway transcription factor GLI1 in Medulloblastoma. Cell Reports. 2015;12(4):599-609. DOI: 10.1016/j.celrep.2015.06.054
  10. 10. Gampala S, Zhang G, Chang CJ, Yang JY. Activation of AMPK sensitizes medulloblastoma to Vismodegib and overcomes Vismodegib-resistance. FASEB Bioadvances. 2021;3(6):459-469. DOI: 10.1096/fba.2020-00032
  11. 11. Hahn H, Wicking C, Zaphiropoulos PG, Gailani MR, Shanley S, Chidambaram A, et al. Mutations of the human homolog of drosophila patched in the nevoid basal cell carcinoma syndrome. Cell. 1996;85:841-851
  12. 12. Johnson RL, Rothman AL, Xie J, Goodrich LV, Bare JW, Bonifas JM, et al. Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science. 1996;272:1668-1671
  13. 13. Amakye D, Jagani Z, Dorsch M. Unraveling the therapeutic potential of the hedgehog pathway in cancer. Nature Medicine. 2013;19(11):1410-1422. DOI: 10.1038/nm.3389
  14. 14. Raffel C, Jenkins RB, Frederick L, Hebrink D, Alderete B, Fults DW, et al. Sporadic Medulloblastomas contain PTCH mutations. Cancer Research. 1997;57:842-845
  15. 15. Xie J, Murone M, Luoh S-M, Ryan A, Gu Q, Zhang C, et al. Activating smoothened mutations in sporadic basal-cell carcinoma. Nature. 1998;391:90-92
  16. 16. Taylor MD, Liu L, Raffel C, Hui C-C, Mainprize TG, Zhang X, et al. Mutations in SUFU predispose to medulloblastoma. Nature Genetics. 2002;31:306-310
  17. 17. Theunissen J-W, de Sauvage FJ. Paracrine hedgehog signaling in cancer. Cancer Research. 2009;69:6007-6010
  18. 18. Truffi M, Sorrentino L, F. Corsi fibroblasts in the tumor microenvironment. Advances in Experimental Medicine and Biology. 2020;1234:15-29
  19. 19. Nicole M, Anderson M, Simon C. The tumor microenvironment. Current Biology. 2020;30(16):R921-R925. DOI: 10.1016/j.cub.2020.06.081
  20. 20. Gampala S, Yang JY. Hedgehog pathway inhibitors against tumor microenvironment. Cell. 2021;10(11):3135. DOI: 10.3390/cells10113135
  21. 21. Onishi H, Nakamura K, Yanai K, Nagai S, Nakayama K, Oyama Y, et al. Cancer therapy that targets the hedgehog signaling pathway considering the cancer microenvironment (review). Oncology Reports. 2022;47(5):93. DOI: 10.3892/or.2022.8304
  22. 22. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nature Medicine. 2018;24(5):541-550. DOI: 10.1038/s41591-018-0014-x
  23. 23. Shan F, Somasundaram A, Bruno TC, Workman CJ, Vignali DAA. Therapeutic targeting of regulatory T cells in cancer. Trends Cancer. 2022;S2405-8033(22):00136-00134. DOI: 10.1016/j.trecan.2022.06.008
  24. 24. O'Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nature Reviews. Clinical Oncology. 2019;16(3):151-167. DOI: 10.1038/s41571-018-0142-8
  25. 25. Gate D, Danielpour M, Rodriguez J Jr, Kim G-B, Levy R, Bannykh S, et al. T-cell TGF-β signaling abrogation restricts medulloblastoma progression. Proceedings of the National Academy of Sciences. 2014;111:E3458-E3466. DOI: 10.1073/pnas.1412489111
  26. 26. Bockmayr M, Mohme M, Klauschen F, Winkler B, Budczies J, Rutkowski S, et al. Subgroup-specific immune and stromal microenvironment in medulloblastoma. Oncoimmunology. 2018;7:e1462430. DOI: 10.1080/2162402X.2018.1462430
  27. 27. Vermeulen JF, Van Hecke W, Adriaansen EJM, Jansen MK, Bouma RG, Villacorta Hidalgo J, et al. Prognostic relevance of tumor-infiltrating lymphocytes and immune checkpoints in pediatric medulloblastoma. Oncoimmunology. 2018;7:e1398877. DOI: 10.1080/2162402X.2017.1398877
  28. 28. Grabovska Y, Mackay A, O’Hare P, Crosier S, Finetti M, Schwalbe EC, et al. Pediatric pan-central nervous system tumor analysis of immune-cell infiltration identifies correlates of antitumor immunity. Nature Communications. 2020;11:4324. DOI: 10.1038/s41467-020-18070-y
  29. 29. Omland SH, Nielsen PS, Gjerdrum LM, Gniadecki R. Immunosuppressive environment in basal cell carcinoma: The role of regulatory T cells. Acta Dermato-Venereologica. 2016;96:917-921. DOI: 10.2340/00015555-2440
  30. 30. Furler RL, Uittenbogaart CH. GLI2 regulates TGF-beta1 in human CD4+ T cells: Implications in cancer and HIV pathogenesis. PLoS One. 2012;7:e40874. DOI: 10.1371/journal.pone.0040874
  31. 31. Eisemann T, Wechsler-Reya RJ. Genes & Development. 2022;36(9–10):514-532
  32. 32. Fan Q, Gu D, Liu H, Yang L, Zhang X, Yoder MC, et al. Defective TGF- beta signaling in bone marrow-derived cells prevents hedgehoginduced skin tumors. Cancer Research. 2014;74:471-483. DOI: 10.1158/0008-5472.CAN-13-2134-T
  33. 33. Fan Q, He M, Sheng T, Zhang X, Sinha M, Luxon B, et al. Requirement of TGFbeta signaling for SMO-mediated carcinogenesis. The Journal of Biological Chemistry. 2010;285:36570-36576. DOI: 10.1074/jbc.C110.164442
  34. 34. Margol AS, Robison NJ, Gnanachandran J, Hung LT, Kennedy RJ, Vali M, et al. Tumor-associated macrophages in SHH subgroup of medulloblastomas. Clinical Cancer Research. 2015;21:1457-1465. DOI: 10.1158/1078-0432.CCR-14-1144
  35. 35. Dang MT, Gonzalez MV, Gaonkar KS, Rathi KS, Young P, Arif S, et al. Macrophages in SHH subgroup medulloblastoma display dynamic heterogeneity that varies with treatment modality. Cell Reports. 2021;34:108917. DOI: 10.1016/j.celrep.2021.108917
  36. 36. Tan IL, Arifa RDN, Rallapalli H, Kana V, Lao Z, Sanghrajka RM, et al. CSF1R inhibition depletes tumor- associated macrophages and attenuates tumor progression in a mouse sonic hedgehog-Medulloblastoma model. Oncogene. 2021;40:396-407. DOI: 10.1038/s41388-020-01536-0
  37. 37. Abad C, Nobuta H, Li J, Kasai A, Yong WH, Waschek JA. Targeted STAT3 disruption in myeloid cells alters immunosuppressor cell abundance in a murine model of spontaneous medulloblastoma. Journal of Leukocyte Biology. 2014;95:357-367. DOI: 10.1189/jlb.1012531
  38. 38. Pham CD, Flores C, Yang C, Pinheiro EM, Yearley JH, Sayour EJ, et al. Differential immune microenvironments and response to immune checkpoint blockade among molecular subtypes of murine medulloblastoma. Clinical Cancer Research. 2016;22:582-595. DOI: 10.1158/1078-0432.CCR-15-0713
  39. 39. Hanna A, Metge BJ, Bailey SK, Chen D, Chandrashekar DS, Varambally S, et al. Inhibition of hedgehog signaling reprograms the dysfunctional immune microenvironment in breast cancer. Oncoimmunology. 2019;8:1548241. DOI: 10.1080/2162402X.2018.1548241
  40. 40. Hill SA, Blaeser AS, Coley AA, Xie Y, Shepard KA, Harwell CC, et al. Sonic hedgehog signaling in astrocytes mediates cell type-specific synaptic organization. eLife. 2019;8:e45545. DOI: 10.7554/eLife.45545
  41. 41. Gingrich EC, Case K, Garcia ADR. A subpopulation of astrocyte progenitors defined by sonic hedgehog signaling. Neural Development. 2022;17(1):2. DOI: 10.1186/s13064-021-00158-w
  42. 42. Xie Y, Kuan AT, Wang W, Herbert ZT, Mosto O, Olukoya O, et al. Astrocyte-neuron crosstalk through hedgehog signaling mediates cortical synapse development. Cell Reports. 2022;38(8):110416. DOI: 10.1016/j.celrep.2022.110416
  43. 43. Liu Y, Yuelling LW, Wang Y, Du F, Gordon RE, O'Brien JA, et al. Astrocytes promote Medulloblastoma progression through hedgehog secretion. Cancer Research. 2017;77(23):6692-6703
  44. 44. Yao M, Ventura PB, Jiang Y, Rodriguez FJ, Wang L, Perry JSA, et al. Astrocytic trans-differentiation completes a multicellular paracrine feedback loop required for Medulloblastoma tumor growth. Cell. 2020;180(3):502-520.e19
  45. 45. Liu H, Sun Y, O’Brien JA, Franco-Barraza J, Qi X, Yuan H, et al. Necroptotic astrocytes contribute to maintaining stemness of disseminated medulloblastoma through CCL2 secretion. Neuro-Oncology. 2020;22:625-638. DOI: 10.1093/neuonc/noz214
  46. 46. Guo D, Wang Y, Cheng Y, Liao S, Hu J, Du F, et al. Tumor cells generate astrocyte-like cells that contribute to SHH-driven medulloblastoma relapse. The Journal of Experimental Medicine. 2021;218(9):e20202350. DOI: 10.1084/jem.20202350
  47. 47. Garzia L, Kijima N, Morrissy AS, De Antonellis P, Guerreiro Stucklin A, Holgado BL, et al. A hematogenous route for medulloblastoma leptomeningeal metastases. Cell. 2018;172:1050-1062.e14. DOI: 10.1016/j.cell.2018.01.038
  48. 48. Saw PE, Chen J, Song E. Targeting CAFs to overcome anticancer therapeutic resistance. Trends Cancer. 2022;8(7):527-555. DOI: 10.1016/j.trecan.2022.03.001
  49. 49. Lavie D, Ben-Shmuel A, Erez N, Scherz-Shouval R. Cancer-associated fibroblasts in the single-cell era. Nature Cancer. 2022;3(7):793-807. DOI: 10.1038/s43018-022-00411-z
  50. 50. Yoshida GJ. Regulation of heterogeneous cancer-associated fibroblasts: The molecular pathology of activated signaling pathways. Journal of Experimental & Clinical Cancer Research. 2020;39(1):112. DOI: 10.1186/s13046-020-01611-0
  51. 51. Thayer SP, di Magliano MP, Heiser PW, Nielsen CM, Roberts DJ, Lauwers GY, et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature. 2003;425(6960):851-856
  52. 52. Berman DM, Karhadkar SS, Maitra A, Montes de Oca R, Gerstenblith MR, Briggs K, et al. Widespread requirement for hedgehog ligand stimulation in growth of digestive tract tumours. Nature. 2003;425:846-851
  53. 53. Bailey JM, Swanson BJ, Hamada T, Eggers JP, Singh PK, Caffery T, et al. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clinical Cancer Research. 2008;14:5995-6004
  54. 54. Feldmann G, Fendrich V, McGovern K, Bedja D, Bisht S, Alvarez H, et al. An orally bioavailable small-molecule inhibitor of hedgehog signaling inhibits tumor initiation and metastasis in pancreatic cancer. Molecular Cancer Therapeutics. 2008;7:2725-2735
  55. 55. Feldmann G, Habbe N, Dhara S, Bisht S, Alvarez H, Fendrich V, et al. Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut. 2008;57:1420-1430
  56. 56. Olive KP, Jacobetz MA, Davidson CJ, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457-1461
  57. 57. 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
  58. 58. Lee JJ, Perera RM, Wang H, Wu D-C, Liu XS, Han S, et al. Stromal response to hedgehog signaling restrains pancreatic cancer progression. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:E3091-E3100
  59. 59. Steele NG, Biffi G, Kemp SB, Zhang Y, Drouillard D, Syu L, et al. Inhibition of hedgehog signaling alters fibroblast composition in pancreatic cancer. Clinical Cancer Research. 2021;27(7):2023-2037. DOI: 10.1158/1078-0432.CCR-20-3715
  60. 60. Kim EJ, Sahai V, Abel EV, Griffith KA, Greenson JK, Takebe N, et al. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clinical Cancer Research. 2014;20(23):5937-5945. DOI: 10.1158/1078-0432.CCR-14-1269
  61. 61. Ko AH, LoConte N, Tempero MA, Walker EJ, Kate Kelley R, Lewis S, et al. A phase I study of FOLFIRINOX plus IPI-926, a hedgehog pathway inhibitor, for advanced pancreatic adenocarcinoma. Pancreas. 2016;45:370-375
  62. 62. Quaglio D, Infante P, Di Marcotullio L, Botta B, Mori M. Hedgehog signaling pathway inhibitors: An updated patent review (2015-present). Expert Opinion on Therapeutic Patents. 2020;30(4):235-250. DOI: 10.1080/13543776.2020.1730327
  63. 63. Incardona JP, Gaffield W, Kapur RP, Roelink H. The teratogenic Veratrum alkaloid cyclopamine inhibits sonic hedgehog signal transduction. Development. 1998;125:3553-3562
  64. 64. Berman DM, Karhadkar SS, Hallahan AR, Pritchard JI, Eberhart CG, Watkins DN, et al. Medulloblastoma growth inhibition by hedgehog pathway blockade. Science. 2002;297:1559-1561
  65. 65. Lin TL, Matsui W. Hedgehog pathway as a drug target: Smoothened inhibitors in development. Oncotargets and Therapy. 2012;5:47
  66. 66. Sekulic A, Migden MR, Oro AE, Dirix L, Lewis KD, Hainsworth JD, et al. Efficacy and safety of Vismodegib in advanced basal-cell carcinoma. The New England Journal of Medicine. 2012;366:2171-2179
  67. 67. Sekulic A, Migden MR, Basset-Seguin N, Garbe C, Gesierich A, Lao CD, et al. Long-term safety and efficacy of vismodegib in patients with advanced basal cell carcinoma: Final update of the pivotal ERIVANCE BCC study. BMC Cancer. 2017;17:332
  68. 68. Pan S, Wu X, Jiang J, Gao W, Wan Y, Cheng D, et al. Discovery of NVP-LDE225, a potent and selective smoothened antagonist. ACS Medicinal Chemistry Letters. 2010;1(3):130-134. DOI: 10.1021/ml1000307
  69. 69. Migden MR, Guminski A, Gutzmer R, Dirix L, Lewis KD, Combemale P, et al. Treatment with two different doses of sonidegib in patients with locally advanced or metastatic basal cell carcinoma (BOLT): A multicentre, randomised, double-blind phase 2 trial. The Lancet Oncology. 2015;16(6):716-728. DOI: 10.1016/S1470-2045(15)70100-2
  70. 70. Munchhof MJ, Li Q, Shavnya A, Borzillo GV, Boyden TL, Jones CS, et al. Discovery of PF-04449913, a potent and orally bioavailable inhibitor of smoothened. ACS Medicinal Chemistry Letters. 2012;3:106-111
  71. 71. Cortes JE, Heidel FH, Hellmann A, Fiedler W, Smith BD, Robak T, et al. Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome. Leukemia. 2019;33:379-389
  72. 72. Chang ALS, Oro AE. Initial assessment of tumor regrowth after Vismodegib in advanced basal cell carcinoma. Archives of Dermatology. 2012;148:1324-1325
  73. 73. Jain R, Dubey SK, Singhvi G. The hedgehog pathway and its inhibitors: Emerging therapeutic approaches for basal cell carcinoma. Drug Discovery Today. 2022;27(4):1176-1183. DOI: 10.1016/j.drudis.2021.12.005
  74. 74. Yao CD, Haensel D, Gaddam S, Patel T, Atwood SX, Sarin KY, et al. AP-1 and TGFß cooperativity drives non-canonical hedgehog signaling in resistant basal cell carcinoma. Nature Communications. 2020;11:5079
  75. 75. Whitson RJ, Lee A, Urman NM, Mirza A, Yao CY, Brown AS, et al. Noncanonical hedgehog pathway activation through SRF–MKL1 promotes drug resistance in basal cell carcinomas. Nature Medicine. 2018;24:271-281
  76. 76. Zhao X, Ponomaryov T, Ornell KJ, Zhou P, Dabral SK, Pak E, et al. RAS/MAPK activation drives resistance to Smo inhibition, metastasis, and tumor evolution in Shh pathway–dependent tumors. Cancer Research. 2015;75:3623-3635
  77. 77. Sharpe HJ, Pau G, Dijkgraaf GJ, Gerrit J, Basset-Seguin N, Modrusan Z, et al. Genomic analysis of smoothened inhibitor resistance in basal cell carcinoma. Cancer Cell. 2015;27:327-341
  78. 78. Zhao X, Pak E, Ornell KJ, Pazyra-Murphy MF, Mac Kenzie EL, Chadwick EJ, et al. Transposon screen identifies loss of primary cilia as a mechanism of resistance to SMO inhibitors. Cancer Discovery. 2017;7:1436-1449
  79. 79. Infante P, Mori M, Alfonsi R, et al. Gli1/DNA interaction is a druggable target for hedgehog-dependent tumors. The EMBO Journal. 2015;34:200-217
  80. 80. Atwood SX, Sarin KY, Whitson RJ, Li JR, Kim G, Rezaee M, et al. Smoothened variants explain the majority of drug resistance in basal cell carcinoma. Cancer Cell. 2015;27:342-353. DOI: 10.1016/j.ccell.2015.02.002
  81. 81. Zhang M, Gao L, Ye Y, Li X. Advances in glioma-associated oncogene (GLI) inhibitors for cancer therapy. Investigational New Drugs. 2022;40(2):370-388. DOI: 10.1007/s10637-021-01187-2
  82. 82. The Nobel Prize in Physiology or Medicine. 2018. Available from: https://www.nobelprize.org/prizes/medicine/2018/summary/
  83. 83. Dammeijer F, van Gulijk M, Mulder EE, Lukkes M, Klaase L, van den Bosch T, et al. The PD-1/PD-L1- checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell. 2020;38(5):685-700.e8. DOI: 10.1016/j.ccell.2020.09.001
  84. 84. Chang ALS, Tran DC, Cannon JGD, Li S, Jeng M, Patel R, et al. Pembrolizumab for advanced basal cell carcinoma: An investigator-initiated, proof- of-concept study. Journal of the American Academy of Dermatology. 2019;80(2):564-566. DOI: 10.1016/j.jaad.2018.08.017
  85. 85. Dummer R, Guminksi A, Gutzmer R, Lear JT, Lewis KD, Chang ALS, et al. Long-term efficacy and safety of sonidegib in patients with advanced basal cell carcinoma: 42-month analysis of the phase II randomized, double-blind BOLT study. The British Journal of Dermatology. 2020;182(6):1369-1378. DOI: 10.1111/bjd.18552
  86. 86. FDA approves cemiplimab-rwlc for locally advanced and metastatic basal cell carcinoma. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-cemiplimab-rwlc-locally-advanced-and-metastatic-basal-cell-carcinoma
  87. 87. Stratigos AJ, Sekulic A, Peris K, et al. Cemiplimab in locally advanced basal cell carcinoma after hedgehog inhibitor therapy: An open-label, multi-Centre, single-arm, phase 2 trial. The Lancet Oncology. 2021;22:848-857
  88. 88. Gambichler T, Stricker I, Neid M, Tannapfel A, Susok L. Impressive response to four cemiplimab cycles of a sonidegib-resistant giant basosquamous carcinoma of the midface. Journal of the European Academy of Dermatology and Venereology. 2022;36(6):e490-e492. DOI: 10.1111/jdv.17993
  89. 89. Grund-Gröschke S, Stockmaier G, Aberger F. Hedgehog/GLI signaling in tumor immunity - new therapeutic opportunities and clinical implications. Cell Communication and Signaling: CCS. 2019;17(1):172. DOI: 10.1186/s12964-019-0459-7
  90. 90. de la Roche M, Ritter AT, Angus KL, Dinsmore C, Earnshaw CH, Reiter JF, et al. Hedgehog signaling controls T cell killing at the immunological synapse. Science. 2013;342(6163):1247-1250. DOI: 10.1126/science.1244689

Written By

Xing-Guo Li and Jer-Yen Yang

Submitted: 01 September 2022 Reviewed: 31 October 2022 Published: 24 January 2023