Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 961 word(s) 961 2021-08-26 10:50:03

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Yang, L. Hedgehog Pathway. Encyclopedia. Available online: https://encyclopedia.pub/entry/13961 (accessed on 20 April 2024).
Yang L. Hedgehog Pathway. Encyclopedia. Available at: https://encyclopedia.pub/entry/13961. Accessed April 20, 2024.
Yang, Ling. "Hedgehog Pathway" Encyclopedia, https://encyclopedia.pub/entry/13961 (accessed April 20, 2024).
Yang, L. (2021, September 07). Hedgehog Pathway. In Encyclopedia. https://encyclopedia.pub/entry/13961
Yang, Ling. "Hedgehog Pathway." Encyclopedia. Web. 07 September, 2021.
Hedgehog Pathway
Edit

The hedgehog pathway, which plays a significant role in embryonic development and stem cell regulation, is activated in gastrointestinal cancers. Chemotherapy is widely used in cancer treatment. However, chemoresistance becomes a substantial obstacle in cancer therapy.

the hedgehog pathway chemotherapy resistance gastric cancer colorectal cancer pancreatic cancer

1. Introduction

The hedgehog (HH) pathway plays a crucial role in embryonic development, tissue homeostasis, and carcinogenesis [1][2]. HH ligands activate signaling by binding to receptor patched 1 homolog (PTCH1). In the absence of HH ligands, PTCH1 prevents smoothened (SMO) from transducing a signal to the downstream glioma-associated oncogene homolog (GLI) transcription factors. HH ligands bind to PTCH1, and relieve PTCH1’s inhibition on SMO, allowing SMO to signal downstream effectors GLI, which activates the target genes via specific genomic DNA sequences (TGGGTGGTC) [3][4].

Activation of GLI proteins via the HH–PTCH1–SMO axis is regarded as the canonical HH signaling pathway. In addition to the canonical pathway, some molecules can bypass the ligand-receptor signaling axis to activate GLI, and these types of regulation are regarded as non-canonical HH signaling. Non-canonical HH signaling is found in malignant diseases. KRAS signaling [5][6], transforming growth factor β (TGFβ) [7], AKT [8], protein kinase C (PKC) [9], and SOX2-bromodomain-containing protein 4(BRD4) [10] are reported to regulate HH signaling via non-canonical pathways.

Chemotherapy is widely used in cancer treatment, and significant improvement is achieved in the prognosis of patients. However, not all patients benefit from it. Chemoresistance becomes a substantial obstacle in cancer therapy due to intrinsic resistance, which occurs at the beginning or even before the treatment, or acquired resistance after initial response to treatment, resulting in relapse [11][12]. Platinum, 5-Fluorouracil (5-FU), and gemcitabine are the most commonly used drugs in the chemotherapy of gastric, colorectal, and pancreatic cancers, and the underlined mechanisms of drug resistance have been studied. Mechanisms of chemoresistance include cancer stem cells(CSCs), tumor microenvironment, and ATP-binding cassette (ABC) transporter family proteins [13][14][15].

Our group studied drug resistance in gastrointestinal cancers and found the HH pathway contributes to drug resistance.

2. New Drugs and Therapeutic Strategy

The HH inhibitors vismodegib and sonidegib have been approved by the Food and Drug Administration to treat recurrent, locally advanced basal cell carcinoma (BCC) or metastatic BCC, or for those who are not eligible for surgery or radiotherapy. The efficacy and safety of vismodegib and sonidegib have been reviewed in [16]. Vismodegib and sonidegib are also used in clinical trials for other solid tumors (medulloblastoma, prostate cancer, pancreatic cancer, and small cell lung cancer) and hematologic malignancies (actively reviewed in [17][18]).The results from these clinical trials show that the HH inhibitors only promote treatment efficacy in HH-driven cancers.

Since current therapy is still far from satisfactory, novel drugs and new therapeutic strategies were developed to improve the treatment. Novel HH inhibitors also have been developed. GDC0449 analog MDB5 [19] and GLI1 inhibitor NanoHHI [20] overcame SMO mutation and improved the treatment effect. Other drugs, such as curcumin, sensitized colorectal cancer to chemotherapy through downregulating HH signaling [21], and Dpc [22], ormeloxifene [23], Patched 1-interacting peptide [24], and metformin [75 ] targeted HH signaling was found to reduce the tumor-associated stromal tissue in pancreatic cancers.

Due to dense stromal tissue, chemotherapeutic and targeted drugs, immune cells are hard to get to cancer cells; therefore, targeting stromal cells is a new promising strategy in pancreatic cancers. Since the HH pathway contributes to the development of the dense stromal tissue, several studies combined SMO inhibitors with either cytotoxic chemotherapeutic drugs [25][26][27] or a targeted antibody [28] to increase the delivery of the drugs and promote tumor infiltration of the CD8 T cells. Inhibition of the HH pathway increased intratumoral vasculature density. Some studies found that SMO inhibitors reduced collagen, α-SMA, and GLI-1 expression [25][28]. However, another study found that SMO inhibitor did not decrease the α-SMA-positive fibroblasts and type I collagen in the stroma [26], indicating more studies should be performed to identify the mechanisms how SMO inhibitors increase the delivery of drugs. Furthermore, research suggested that combined the hepatocyte growth factor (HGF)/c-Met and HH pathways inhibitors overcame the resistance to the single-inhibitor treatment and led to sensitization to the gemcitabine treatment [29]. Despite the promising results above and the excellent responses to sonidegib in a mouse model [30], vismodegib does not show improvement in metastatic pancreatic adenocarcinoma (NCT01088815, NCT01064622, [31]). The preclinical model may not accurately reflect the tumor context of patients; patient-derived xenografts, and maybe in the future, patient-derived 3D culture models with tumor cells and a microenvironment, are better materials for studying the efficacy and mechanisms of action of therapeutic drugs.

3. Conclusions and Perspectives

Accumulating data suggest that the HH pathway plays an important role in chemoresistance in gastrointestinal cancers. CSCs are the well-known cause for drug resistance and are extensively studied in gastric, colorectal, and pancreatic cancers, and the HH pathway is a promising target for eradicating CSCs. Due to the dense stromal tissue in pancreatic cancers, the role of HH signaling in PSCs is actively being investigated. Inhibition of the HH pathway in PSCs reduces stromal tissue and increases drug delivery, suggesting that HH signaling may also play a mechanical role in chemoresistance. However, studies focused on the HH pathway in the TME of gastric and colorectal cancer chemoresistance are relatively scarce. Despite the different pathological characteristics in gastric, colorectal, and pancreatic cancers, the HH pathway regulates the ABC transporter family proteins in all three types of cancer.
Studies from gastrointestinal cancers and their CSCs provide evidence for the existence of both canonical and non-canonical HH signaling, which do sound the alarm to us. Inhibition of SMO may not inhibit HH activation, and this may partially explain the dismal results of vismodegib in some clinical trials for advanced solid tumors. Identifying how HH signaling is activated, caused by either a ligand-dependent or ligand-independent mechanism, may help us choose the correct inhibitors to attenuate activation of the HH pathway in different cancer contexts.

References

  1. Kong, J.; Siebold, C.; Rohatgi, R. Biochemical mechanisms of vertebrate hedgehog signaling. Development 2019, 146, dev166892.
  2. Yang, L.; Xie, G.; Fan, Q.; Xie, J. Activation of the hedgehog-signaling pathway in human cancer and the clinical implications. Oncogene 2010, 29, 469–481.
  3. Kinzler, K.; Vogelstein, B. The GLI gene encodes a nuclear protein which binds specific sequences in the human genome. Mol. Cell. Biol. 1990, 10, 634–642.
  4. Sasaki, H.; Hui, C.; Nakafuku, M.; Kondoh, H. A binding site for Gli proteins is essential for HNF-3beta floor plate enhancer activity in transgenics and can respond to Shh in vitro. Development 1997, 124, 1313–1322.
  5. Ji, Z.; Mei, F.; Xie, J.; Cheng, X. Oncogenic KRAS activates hedgehog signaling pathway in pancreatic cancer cells. J. Biol. Chem. 2007, 282, 14048–14055.
  6. Seto, M.; Ohta, M.; Asaoka, Y.; Ikenoue, T.; Tada, M.; Miyabayashi, K.; Mohri, D.; Tanaka, Y.; Ijichi, H.; Tateishi, K.; et al. Regulation of the hedgehog signaling by the mitogen-activated protein kinase cascade in gastric cancer. Mol. Carcinog. 2009, 48, 703–712.
  7. Dennler, S.; André, J.; Alexaki, I.; Li, A.; Magnaldo, T.; ten Dijke, P.; Wang, X.; Verrecchia, F.; Mauviel, A. Induction of sonic hedgehog mediators by transforming growth factor-beta: Smad3-dependent activation of Gli2 and Gli1 expression in vitro and in vivo. Cancer Res. 2007, 67, 6981–6986.
  8. Stecca, B.; Mas, C.; Clement, V.; Zbinden, M.; Correa, R.; Piguet, V.; Beermann, F.; Ruiz i Altaba, A. Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proc. Natl. Acad. Sci. USA 2007, 104, 5895–5900.
  9. Cai, Q.; Li, J.; Gao, T.; Xie, J.; Evers, B. Protein kinase Cdelta negatively regulates hedgehog signaling by inhibition of Gli1 activity. J. Biol. Chem. 2009, 284, 2150–2158.
  10. Pietrobono, S.; Gaudio, E.; Gagliardi, S.; Zitani, M.; Carrassa, L.; Migliorini, F.; Petricci, E.; Manetti, F.; Makukhin, N.; Bond, A.; et al. Targeting non-canonical activation of GLI1 by the SOX2-BRD4 transcriptional complex improves the efficacy of HEDGEHOG pathway inhibition in melanoma. Oncogene 2021, 40, 3799–3814.
  11. Wijdeven, R.; Pang, B.; Assaraf, Y.; Neefjes, J. Old drugs, novel ways out: Drug resistance toward cytotoxic chemotherapeutics. Drug Resist. Updates Rev. Comment. Antimicrob. Anticancer Chemother. 2016, 28, 65–81.
  12. Alexa-Stratulat, T.; Pešić, M.; Gašparović, A.; Trougakos, I.; Riganti, C. What sustains the multidrug resistance phenotype beyond ABC efflux transporters? Looking beyond the tip of the iceberg. Drug Resist. Updates Rev. Comment. Antimicrob. Anticancer Chemother. 2019, 46, 100643.
  13. Martins-Neves, S.; Cleton-Jansen, A.; Gomes, C. Therapy-induced enrichment of cancer stem-like cells in solid human tumors: Where do we stand? Pharmacol. Res. 2018, 137, 193–204.
  14. Katoh, M. Genomic testing, tumor microenvironment and targeted therapy of Hedgehog-related human cancers. Clin. Sci. 2019, 133, 953–970.
  15. Uzunparmak, B.; Sahin, I. Pancreatic cancer microenvironment: A current dilemma. Clin. Transl. Med. 2019, 8, 2.
  16. Migden, M.; Farberg, A.S.; Dummer, R.; Squittieri, N.; Hanke, C.W. A Review of Hedgehog Inhibitors Sonidegib and Vismodegib for Treatment of Advanced Basal Cell Carcinoma. J. Drugs Dermatol. JDD 2021, 20, 156–165.
  17. Cortes, J.E.; Gutzmer, R.; Kieran, M.W.; Solomon, J.A. Hedgehog signaling inhibitors in solid and hematological cancers. Cancer Treat Rev. 2019, 76, 41–50.
  18. Xie, H.; Paradise, B.D.; Ma, W.W.; Fernandez-Zapico, M.E. Recent advances in the clinical targeting of Hedgehog/GLI signaling in cancer. Cells 2019, 8, 394.
  19. Kumar, V.; Chaudhary, A.; Dong, Y.; Zhong, H.; Mondal, G.; Lin, F.; Kumar, V.; Mahato, R. Design, synthesis and biological evaluation of novel hedgehog inhibitors for treating pancreatic cancer. Sci. Rep. 2017, 7, 1665.
  20. Chenna, V.; Hu, C.; Pramanik, D.; Aftab, B.; Karikari, C.; Campbell, N.; Hong, S.; Zhao, M.; Rudek, M.; Khan, S.; et al. A polymeric nanoparticle encapsulated small-molecule inhibitor of Hedgehog signaling (NanoHHI) bypasses secondary mutational resistance to Smoothened antagonists. Mol. Cancer Ther. 2012, 11, 165–173.
  21. Ramasamy, T.; Ayob, A.; Myint, H.; Thiagarajah, S.; Amini, F. Targeting colorectal cancer stem cells using curcumin and curcumin analogues: Insights into the mechanism of the therapeutic efficacy. Cancer Cell Int. 2015, 15, 96.
  22. Geleta, B.; Park, K.; Jansson, P.; Sahni, S.; Maleki, S.; Xu, Z.; Murakami, T.; Pajic, M.; Apte, M.; Richardson, D.; et al. Breaking the cycle: Targeting of NDRG1 to inhibit bi-directional oncogenic cross-talk between pancreatic cancer and stroma. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2021, 35, e21347.
  23. Khan, S.; Ebeling, M.; Chauhan, N.; Thompson, P.; Gara, R.; Ganju, A.; Yallapu, M.; Behrman, S.; Zhao, H.; Zafar, N.; et al. Ormeloxifene suppresses desmoplasia and enhances sensitivity of gemcitabine in pancreatic cancer. Cancer Res. 2015, 75, 2292–2304.
  24. Oyama, Y.; Onishi, H.; Koga, S.; Murahashi, M.; Ichimiya, S.; Nakayama, K.; Fujimura, A.; Kawamoto, M.; Imaizumi, A.; Umebayashi, M.; et al. Patched 1-interacting Peptide Represses Fibrosis in Pancreatic Cancer to Augment the Effectiveness of Immunotherapy. J. Immunother. 2020, 43, 121–133.
  25. Wang, L.; Liu, X.; Zhou, Q.; Sui, M.; Lu, Z.; Zhou, Z.; Tang, J.; Miao, Y.; Zheng, M.; Wang, W.; et al. Terminating the criminal collaboration in pancreatic cancer: Nanoparticle-based synergistic therapy for overcoming fibroblast-induced drug resistance. Biomaterials 2017, 144, 105–118.
  26. Zhao, J.; Xiao, Z.; Li, T.; Chen, H.; Yuan, Y.; Wang, Y.; Hsiao, C.; Chow, D.; Overwijk, W.; Li, C. Stromal modulation reverses primary resistance to immune checkpoint blockade in pancreatic cancer. ACS Nano 2018, 12, 9881–9893.
  27. Olive, K.; Jacobetz, M.; Davidson, C.; Gopinathan, A.; McIntyre, D.; Honess, D.; Madhu, B.; Goldgraben, M.; Caldwell, M.; Allard, D.; et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science 2009, 324, 1457–1461.
  28. Wang, J.; Chan, D.; Sen, A.; Ma, W.; Straubinger, R. Tumor priming by SMO inhibition enhances antibody delivery and efficacy in a pancreatic ductal adenocarcinoma model. Mol. Cancer Ther. 2019, 18, 2074–2084.
  29. Rucki, A.; Xiao, Q.; Muth, S.; Chen, J.; Che, X.; Kleponis, J.; Sharma, R.; Anders, R.; Jaffee, E.; Zheng, L. Dual inhibition of hedgehog and c-Met pathways for pancreatic cancer treatment. Mol. Cancer Ther. 2017, 16, 2399–2409.
  30. Fendrich, V.; Wiese, D.; Waldmann, J.; Lauth, M.; Heverhagen, A.E.; Rehm, J.; Bartsch, D.K. Hedgehog inhibition with the orally bioavailable Smo antagonist LDE225 represses tumor growth and prolongs survival in a transgenic mouse model of islet cell neoplasms. Ann. Surg. 2011, 254, 818–823, discussion 823.
  31. McCleary-Wheeler, A.L.; Carr, R.M.; Palmer, S.R.; Smyrk, T.C.; Allred, J.B.; Almada, L.L.; Tolosa, E.J.; Lamberti, M.J.; Marks, D.L.; Borad, M.J.; et al. Phase 1 trial of Vismodegib and Erlotinib combination in metastatic pancreatic cancer. Pancreatol. Off. J. Int. Assoc. Pancreatol. 2020, 20, 101–109.
More
Information
Subjects: Oncology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 327
Revision: 1 time (View History)
Update Date: 07 Sep 2021
1000/1000