Open Access

Calcitonin gene‑related peptide: A promising bridge between cancer development and cancer‑associated pain in oral squamous cell carcinoma (Review)

  • Authors:
    • Yu Zhang
    • Chengzhong Lin
    • Xu Wang
    • Tong Ji
  • View Affiliations

  • Published online on: September 18, 2020     https://doi.org/10.3892/ol.2020.12116
  • Article Number: 253
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Nerves have been widely demonstrated to exert major effects in tumor‑associated microenvironments. Due to the characteristic innervation of the oral cavity and the fact that cancer‑associated pain is a distinct feature of oral squamous cell carcinoma (OSCC), the sensory nerves may dominate in the OSCC‑nerve microenvironment. As the most abundant neuropeptide in the trigeminal ganglion, the calcitonin gene‑related peptide (CGRP) exerts a dual effect on cancer development and cancer‑associated pain in various types of cancer. The present review explored the potential molecular mechanisms of the roles of CGRP in cancer development and cancer‑associated pain, suggesting that CGRP may be a promising therapeutic target for OSCC.

Introduction

Similarly to normal organs, tumors establish their own microenvironment to fulfill their dynamic growing demands. This microenvironment may include the extracellular matrix, vessels, lymphangions and, as an increasingly recognized compartment, nerves (1). The role of vessels in tumor microenvironments has been widely explored (2,3). However, so for the curative effects of anti-angiogenesis treatments have been proven dissatisfying, and more and more studies find that the nerves play a pivotal role in various cancer (4,5).

Oral cancer is a common malignant cancer type in the head and neck region with over 50,000 new cases and 10,000 new deaths every year in the U.S.A (6). Oral squamous cell carcinoma (OSCC) constitutes 90% of cases of oral cancer (6). OSCC is associated with severe disease and treatment-associated morbidity, and is frequently reported as having high rates of recurrence despite advances in cancer treatment (7). The oral cavity is innervated by cranial nerves with a high density of sensory nerves, particularly the trigeminal nerve (8). Therefore, pain is the most frequent complaint at the primary site (9), and the prevalence and intensity of OSCC-associated pain are higher than those in all other types of cancer (10,11). The mechanisms involved in the OSCC-nerve microenvironment are likely to differ from those of prostate, pancreatic and breast cancer, which are predominantly innervated by autonomic nerves (4,12).

As the most abundant neuropeptide in the trigeminal ganglion, the calcitonin gene-related peptide (CGRP) exerts a dual effect on both cancer development and cancer-associated pain in various types of cancer, such as osteosarcoma (13) and breast cancer (14). The present review discussed the potential molecular mechanisms by which CGRP is implicated in the development of OSCC and associated pain, suggesting that CGRP may be a promising therapeutic target in OSCC (Fig. 1).

CGRP and its inhibitors

CGRP is a 37-amino acid peptide that has a ubiquitous distribution, particularly in trigeminal ganglions (TGs) (15). CGRP has two major isoforms: α- and β-CGRP, which have similar structures and biological activities, but are encoded by separate genes(CALCA for α-CGRP and CALCB for β-CGRP) (16). In general, it is thought that α-CGRP is the cardinal form occurring in the central and peripheral nervous system, whereas β-CGRP, the only product of the CALCB gene, acts mainly in the enteric nervous system (16). Along with the wide innervation of C and Aδ fibers, CGRP exerts various physical effects, including the regulation of energy metabolism, cardiovascular function and pain conduction (17). CGRP has crucial functions in pathological situations, including hypertension, migraine, atherosclerosis and vessel remodeling, which has been elaborated on in a previous review (17).

The CGRP receptor is present in both central and peripheral nerves (18). It is composed of three proteins, namely the calcitonin receptor-like receptor (CLR), receptor activity-modifying protein 1 (RAMP1) and receptor component protein (RCP) (18). The CLR belongs to the class B ‘secretin-like’ family of G-protein-coupled receptors (GPCRs) (18). RAMP1s are small membrane proteins that possess a large extracellular NH2 terminus of 100 amino acids, a single transmembrane domain and a short intracellular domain of 10 amino acids (19). RCP is a 16-kDa protein that interacts with the second intracellular cytoplasmic loop of the CLR (20). It is hypothesized that the CLR, RAMP1 and RCP compose a high-affinity receptor complex for CGRP that may be antagonized by the short peptide CGRP8-37 and non-peptide antagonists, such as rimegepant (21).

With the revelation of the pivotal role of the CGRP/CLR axis in migraines (22), successful therapeutic strategies using humanized monoclonal antibodies or non-peptide antagonists directed against CGRP or CLR have emerged. Antibodies against CGRP, such as fremanezumab (23) and galcanezumab (24), or against CLR, such as erenumab (25), have been approved by the Food and Drug Administration (FDA) for the treatment of migraine. Non-peptide antagonists of CLR, such as ubrogepant (26,27), were also approved by the FDA, and rimegepant has been through phase III clinical trials (28,29) for the treatment of migraine. These antibodies and antagonists have been useful for the study of the CGRP/CLR axis.

CGRP and cancer-associated pain

Pain is one of the most common factors affecting the quality of life in patients with cancer and 75–95% of patients with metastatic or advanced-stage cancer will experience considerable amounts of cancer-induced pain (30). The incidence rate of cancer related pain varies for different types of cancers. For instance, breast adenocarcinoma is frequently characterized as being painful only after it metastasizes to the bone (31). However, melanomas (even in the head and neck) are typically not painful at the primary site or metastatic sites (32). In OSCC, pain is the most frequent complaint at the primary site (9), and the prevalence and intensity of OSCC-associated pain are higher than in all other types of cancer (10,11). In a prospective study, the pretreatment pain in OSCC has been able to predict peripheral nerve invasion (33), and was identified as an independent predictor of survival in 2,340 patients with OSCC in a retrospective study (10). The dorsal root ganglion (DRG) and TG neurons are pseudounipolar, which means that the axon bifurcates and sends projections to both the dorsal horn of the spinal cord or brainstem, and to the periphery (34). This unique morphology allows the release of transmitters at central and peripheral sites, and allows for bidirectional communication between the two terminals (35). When peripheral noxious stimuli (including high hydrogen, high temperature, capsaicin or prostaglandin E2) act on peripheral nociceptors, CGRP serves a dual role in afferent and efferent nerves (36). On one hand, CGRP is released in the synapse of primary afferent nerves through anterograde axoplasmic transport and acts on its receptors on the postsynaptic neurons in the DRG and TG, thus transmitting algesia to pain centers. On the other hand, it is also released from peripheral nerve terminals and exerts paracrine effects on the surrounding tissues by retrograde axoplasmic transport (36).

CGRP modulates pain in two major ways. A relatively well-researched mechanism is that following the vasodilation effect of CGRP, the accumulated proinflammatory factors irritate peripheral nociceptors. In addition, CGRP may directly influence peripheral and central sensitization, resulting in hyperalgesia and allodynia (37) (Fig. 2). A recent study demonstrated that CGRP induces differential regulation of cytokines, mostly interleukin (IL)-1β and IL-6, from satellite glial cells in TG, causing increased pain (37). In most situations, these two effects often co-exist and mutually promote each other (36).

The present review summarized the existent studies on the role of CGRP in cancer-associated pain (Table SI). Most studies used in vivo experiments to reveal that there are higher numbers of CGRP(+) sensory nerves sprouting adjacent to an ectopic cancer and in the DRG or TG than elsewhere, and mechanical allodynia and thermal hyperalgesia exist in various types of tumor, such as gingival cancer, metastatic bone cancer and breast cancer (35,3845). Animal models should be optimized to study the role of CGRP in OSCC-associated pain. Nagamine et al (44) established a rat model of oral cancer pain by inoculating cancer cells into the lower gingiva, and instead of hind paw withdrawal, used head withdrawal as the evaluation index; in addition, it was revealed that the mechanical allodynia and thermal hyperalgesia in the ipsilateral maxillary and mandibular nerve area were accompanied by nerves with upregulated CGRP expression in the TG. Although this was an innovative discovery, perhaps adding another assessment index, such as food intake, may help to confirm this result.

Overall, CGRP exerts considerable roles in cancer-associated pain, as well as in OSCC. The active sites of CGRP are the peripheral nociceptors and central nerve endings in the DRG or TG. However, the current in vivo models and antagonists require further improvements.

CGRP in cancer development

Initially, high levels of CGRP were identified in tumor tissues and serum of patients with medullary thyroid carcinoma (46). Subsequent studies emphasized the diagnostic value of CGRP in non-small cell lung carcinoma (47,48), neuroendocrine tumors (49), prostate cancer (50,51) and particularly medullary thyroid carcinoma (46,52). Recently, Angenendt et al (53) analyzed the expression levels of the gene encoding the CLR, CALCRL, in >1,500 patients with well-characterized acute myeloid leukemia from five international cohorts, revealing that increasing transcript levels of CALCRL were associated with decreasing complete remission rates, 5-year overall survival and event-free survival. Furthermore, CRISPR-Cas9-mediated knockout of CALCRL markedly impaired colony formation in human myeloid leukemia cell lines (53).

One way in which CGRP is able to influence tumor behavior is secondary to its potent vasodilation effect. A previous study on CGRP-knockout mice revealed that endogenous CGRP facilitated tumor-associated angiogenesis and tumor growth in Lewis lung carcinoma-bearing mice and that the administration of the CGRP antagonist CGRP8-37 or denervation markedly suppressed tumor growth and tumor-associated angiogenesis (54).

A number of studies have revealed that CGRP can directly modulate development of various types of cancer, with a large amount of literature reporting the expression of the CGRP receptor complex in cancer cells (5557). CGRP was demonstrated to be able to modify the chemokinetic abilities of a metastatic breast cancer cell line and increase the expression of its receptors (58). In addition, it has been demonstrated that CGRP increased the invasive ability of prostate cancer PC-3 cells and an osteosarcoma cell line (13,50). Recently, Dallmayer et al (56) revealed that targeting the CALCB/RAMP1 (the receptor complex of β-CGRP) axis inhibited the growth of Ewing sarcoma cells. Despite the lack of studies on the role of CGRP in OSCC, several studies have revealed a wide distribution of CLR/RAMP1 in oral soft tissues, bones and dental tissues (59). Therefore, the present review discussed the mechanisms that CGRP may utilize in cancer development.

Aerobic glycolysis

It is well known that rapidly growing cancer cells tend to utilize glycolysis during proliferation, regardless of oxygen availability, which is known as the Warburg effect (60). Aerobic glycolysis is one of the hallmarks of cancer, including OSCC (61). Various enzymes participating in glycolysis are upregulated in OSCC and are associated with poor prognosis (6264). Rossetti et al (65) demonstrated that CGRP potently antagonizes the effect of insulin in glycogen synthesis and enhances glycolysis in muscles, which was attributed to the activation of cyclic adenosine monophosphate (cAMP). However, a recent study on insulin resistance revealed that with the activation of transient receptor potential vanilloid-1 proteins/CGRP axis, glucose transporter 4 expression was upregulated at the protein and mRNA levels, which ameliorated insulin resistance (66). Despite the disparity of certain results, these findings suggest that CGRP may serve important roles in cell glycometabolism.

A pivotal signaling pathway in cell glycometabolism in which CGRP participates is the AMP-activated protein kinase (AMPK) signaling pathway (67). AMPK senses the intracellular AMP/adenosine triphosphate (ATP) or adenosine diphosphate/ATP levels, promoting metabolic pathways that generate ATP, such as glycolysis and lipolysis, and inhibiting those consuming ATP, such as glycogen and lipid synthesis (68). Danaher et al (67) suggested that CGRP activates AMPK through phosphorylation of Thr172 in its α-subunit. As a serine/threonine protein kinase that works as a sensor of cellular energy, phosphorylated AMPK is able to directly phosphorylate proteins or modulate gene transcription involved in glucose and lipid metabolism, which increases the cell utilization of glucose (68).

Lipid metabolism

Lipid metabolism is also reprogrammed in different types of cancer, but not in the same way for all. Multiple lines of evidence suggest that CGRP may regulate lipid metabolism in physiological and pathological conditions. Compared with normal-weight females, the plasma CGRP concentration is increased in obese females (body mass index >35 kg/m2) (69). In pre-obese Zucker rats, the fasting plasma CGRP levels were elevated in lean animals prior to the appearance of body weight differences compared to control group (70). Furthermore, CGRP-knockout mice were observed to be protected from diet-induced obesity compared with CGRP+/+ mice, without any obvious attenuation of food intake (70). Danaher et al (67) suggested that levels of CGRP similar to those in the blood markedly stimulated fatty acid β-oxidation and evoked concomitant mobilization of muscle lipid via receptor-mediated activation of muscle lipolysis in rodents. In OSCC, it was demonstrated that fenofibrate, a particularly potent clinical lipid-lowering agent, suppressed oral tumorigenesis and cancer development by downregulating mTOR activity and activating the AMPK signaling pathway (71). In another retrospective study including 576 patients diagnosed with T1/2N0M0 OSCC without any weight loss prior to diagnosis, the progression-free survival time was poorer in obese patients than in those of normal weight (72). Future studies should investigate whether CGRP may regulate lipid metabolism in OSCC.

Aberrant internalization and intracellular trafficking

It is normal for the internalization of receptors to serve as a physiological feedback mechanism to switch off the initiated cellular signaling pathways. However, studies on certain GPCR-like receptors, such as the thyroid-stimulating hormone (TSH) receptor and the parathyroid hormone receptor, have demonstrated that an internalized ligand-receptor compound is able to convey sustained signals in endosomes (73,74). The CLR/RAMP1 receptor complex is internalized together into an endosome after combining with CGRP (75). Yarwood et al (76) revealed that endosomal CLR is able to activate protein kinase C in the cytosol, as well as ERK in the cytosol and nucleus. In addition, the authors developed a cholestanol-conjugated antagonist (CGRP8-37-cholestanol) to prevent sustained neuronal excitation caused by CGRP, which accumulates in endosomes and is capable of inhibiting endosomal CLR/cAMP signaling without affecting CLR/RAMP1 receptors at the cell surface. A similar result was obtained in a study on substance P, which is frequently regarded as coexisting with CGRP (77,78). A number of studies agree that endosomal signaling is more efficient compared with the traditional plasma membrane signaling and may result in tumorigenesis (76,79). Godbole et al (79) demonstrated that the TSH receptor co-internalizes with TSH and traffics retrogradely to the trans-Golgi network, where it activates an endogenous pool of Gs proteins; this leads to a delayed phase of local cAMP production and protein kinase A activation at a critical position near the nucleus, which appears to be required for efficient phosphorylation and gene transcription of the cAMP response element-binding protein.

Overall, the aberrant internalization of receptors contributes to tumorigenesis. CLR/RAMP1 may be sustained and activated through an endosomal signaling pathway, which is an unexploited but promising field in OSCC. Cholestanol-conjugated antagonists may represent a novel approach for receptor antagonist development.

Immune evasion

Immune evasion is another hallmark of cancer (80). Previous studies have demonstrated that the aforementioned effects may be at least partially mediated by CGRP. Anatomically, the peripheral nervous system is connected with lymphoid organs through sensory nerves (81,82). In addition to sensory nerves, CGRP may be produced by immune cells, including T cells (83,84), B cells (85) and monocytes (86). The CLR/RAMP1 receptor appears to be expressed by most immune cells (87,88). Endogenous CGRP from nerves or immune cells is a major suppressor of immune reactions (89,90). In differentiated CD4+ T cells, CGRP inhibits the production of tumor necrosis factor-α and interferon-γ by T helper type 1 cells via elevating intracellular cAMP levels, while IL-4 production by T helper type 2 cells is not affected (89,90). Following treatment of Langerhans cells with CGRP, the capacity of these cells to stimulate the proliferation of murine T cells was impaired (91). Similarly, treating human monocytes or dendritic cells with CGRP markedly compromised the proliferative response of allogeneic T cells through the release of IL-10 (92). The immunomodulatory roles of CGRP are explained more in detail in a previous review (93).

In OSCC, an impaired and suppressed immune function in both the whole human system and in the local tumor microenvironment is associated with poor prognosis for patients with OSCC (61). Future studies should analyze how CGRP may regulate immune reactions in OSCC.

Conclusion and future directions

Clinical and pathological features confirm that nerves serve an important role in OSCC, as well as in other types of cancer. However, sensory nerves, rather than autonomic nerves, appear to have a pivotal role in OSCC. CGRP is mostly expressed in sensory neurons in the TG and DRG (94,95). CGRP exerts a significant role in both cancer development and cancer-associated pain. CGRP promotes cancer development through metabolic reprogramming, anomalous receptor internalization or inhibition of antitumor immune responses. In addition, CGRP may augment pain by inducing mechanical allodynia peripherally and centrally. Based on the current knowledge, the present review suggested that CGRP may represent an important bridge between cancer development and cancer-associated pain in OSCC, and this hypothesis should be further investigated in future studies.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present review was supported by the National Natural Science Foundation of China (grant nos. 8187100500 and 81702675)) and the Science and Technology Commission of Shanghai Municipality (grant no. YDZX20173100004422).

Availability of data and materials

Not applicable.

Authors' contributions

YZ, TJ and XW conceived and designed the study. CL researched the literature. YZ wrote the manuscript. CL and XW designed the figures. YZ, CL, XW and TJ revised and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

OSCC

oral squamous cell carcinoma

CGRP

calcitonin gene-related peptide

TG

trigeminal ganglions

CLR

calcitonin receptor-like receptor

RAMP1

receptor activity-modifying protein 1

RCP

receptor component protein

GPCR

G-protein-coupled receptor

DRG

dorsal root ganglion

AMPK

adenosine monophosphate-activated protein kinase

References

1 

Senthebane DA, Rowe A, Thomford NE, Shipanga H, Munro D, Mazeedi MAMA, Almazyadi HAM, Kallmeyer K, Dandara C, Pepper MS, et al: The role of tumor microenvironment in chemoresistance: To survive, keep your enemies closer. Int J Mol Sci. 18:15862017. View Article : Google Scholar

2 

Yadav L, Puri N, Rastogi V, Satpute P and Sharma V: Tumour angiogenesis and angiogenic inhibitors: A review. J Clin Diagn Res. 9:XE01–XE05. 2015.PubMed/NCBI

3 

Boilly B, Faulkner S, Jobling P and Hondermarck H: Nerve dependence: From regeneration to cancer. Cancer Cell. 31:342–354. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Amit M, Takahashi H, Dragomir MP, Lindemann A, Gleber-Netto FO, Pickering CR, Anfossi S, Osman AA, Cai Y, Wang R, et al: Loss of p53 drives neuron reprogramming in head and neck cancer. Nature. 578:449–454. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Mauffrey P, Tchitchek N, Barroca V, Bemelmans AP, Firlej V, Allory Y, Roméo PH and Magnon C: Progenitors from the central nervous system drive neurogenesis in cancer. Nature. 569:672–678. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2018. CA Cancer J Clin. 68:7–30. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Rogers SN, Brown JS, Woolgar JA, Lowe D, Magennis P, Shaw RJ, Sutton D, Errington D and Vaughan D: Survival following primary surgery for oral cancer. Oral Oncol. 45:201–211. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Mu L and Sanders I: Human tongue neuroanatomy: Nerve supply and motor endplates. Clin Anat. 23:777–791. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Bagan J, Sarrion G and Jimenez Y: Oral cancer: Clinical features. Oral Oncol. 46:414–417. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Reyes-Gibby CC, Anderson KO, Merriman KW, Todd KH, Shete SS and Hanna EY: Survival patterns in squamous cell carcinoma of the head and neck: Pain as an independent prognostic factor for survival. J Pain. 15:1015–1022. 2014. View Article : Google Scholar : PubMed/NCBI

11 

van den Beuken-van Everdingen MH, de Rijke JM, Kessels AG, Schouten HC, van Kleef M and Patijn J: Prevalence of pain in patients with cancer: A systematic review of the past 40 years. Ann Oncol. 18:1437–1449. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Zahalka AH and Frenette PS: Nerves in cancer. Nat Rev Cancer. 20:143–157. 2020. View Article : Google Scholar : PubMed/NCBI

13 

Drissi H, Lieberherr M, Hott M, Marie PJ and Lasmoles F: Calcitonin gene-related peptide (CGRP) increases intracellular free Ca2+ concentrations but not cyclic AMP formation in CGRP receptor-positive osteosarcoma cells (OHS-4). Cytokine. 11:200–207. 1999. View Article : Google Scholar : PubMed/NCBI

14 

Papantoniou V, Tsiouris S, Sotiropoulou M, Valsamaki P, Koutsikos J, Ptohis N, Dimitrakakis C, Sotiropoulou E, Melissinou M, Nakopoulou L, et al: The potential role of calcitonin gene-related peptide (CGRP) in breast carcinogenesis and its correlation with 99mTc-(V)DMSA scintimammography. Am J Clin Oncol. 30:420–427. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Eftekhari S, Salvatore CA, Calamari A, Kane SA, Tajti J and Edvinsson L: Differential distribution of calcitonin gene-related peptide and its receptor components in the human trigeminal ganglion. Neuroscience. 169:683–696. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Brain SD and Grant AD: Vascular actions of calcitonin gene-related peptide and adrenomedullin. Physiol Rev. 84:903–934. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Russell FA, King R, Smillie SJ, Kodji X and Brain SD: Calcitonin gene-related peptide: Physiology and pathophysiology. Physiol Rev. 94:1099–1142. 2014. View Article : Google Scholar : PubMed/NCBI

18 

Rosenfeld MG, Mermod JJ, Amara SG, Swanson LW, Sawchenko PE, Rivier J, Vale WW and Evans RM: Production of a novel neuropeptide encoded by the calcitonin gene via tissue-specific RNA processing. Nature. 304:129–135. 1983. View Article : Google Scholar : PubMed/NCBI

19 

McLatchie LM, Fraser NJ, Main MJ, Wise A, Brown J, Thompson N, Solari R, Lee MG and Foord SM: RAMPs regulate the transport and ligand specificity of the calcitonin-receptor-like receptor. Nature. 393:333–339. 1998. View Article : Google Scholar : PubMed/NCBI

20 

Luebke AE, Dahl GP, Roos BA and Dickerson IM: Identification of a protein that confers calcitonin gene-related peptide responsiveness to oocytes by using a cystic fibrosis transmembrane conductance regulator assay. Proc Natl Acad Sci USA. 93:3455–3460. 1996. View Article : Google Scholar : PubMed/NCBI

21 

Negro A and Martelletti P: Gepants for the treatment of migraine. Expert Opin Investig Drugs. 28:555–567. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Edvinsson L, Haanes KA, Warfvinge K and Krause DN: CGRP as the target of new migraine therapies-successful translation from bench to clinic. Nat Rev Neurol. 14:338–350. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Ferrari MD, Diener HC, Ning X, Galic M, Cohen JM, Yang R, Mueller M, Ahn AH, Schwartz YC, Grozinski-Wolff M, et al: Fremanezumab versus placebo for migraine prevention in patients with documented failure to up to four migraine preventive medication classes (FOCUS): A randomised, double-blind, placebo-controlled, phase 3b trial. Lancet. 394:1030–1040. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Goadsby PJ, Dodick DW, Leone M, Bardos JN, Oakes TM, Millen BA, Zhou C, Dowsett SA, Aurora SK, Ahn AH, et al: Trial of galcanezumab in prevention of episodic cluster headache. N Engl J Med. 381:132–141. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Reuter U, Goadsby PJ, Lanteri-Minet M, Wen S, Hours-Zesiger P, Ferrari MD and Klatt J: Efficacy and tolerability of erenumab in patients with episodic migraine in whom two-to-four previous preventive treatments were unsuccessful: A randomised, double-blind, placebo-controlled, phase 3b study. Lancet. 392:2280–2287. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Dodick DW, Lipton RB, Ailani J, Lu K, Finnegan M, Trugman JM and Szegedi A: Ubrogepant for the treatment of migraine. N Engl J Med. 381:2230–2241. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Lipton RB, Dodick DW, Ailani J, Lu K, Finnegan M, Szegedi A and Trugman JM: Effect of ubrogepant vs placebo on pain and the most bothersome associated symptom in the acute treatment of migraine: The ACHIEVE II randomized clinical trial. JAMA. 322:1887–1898. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Lipton RB, Croop R, Stock EG, Stock DA, Morris BA, Frost M, Dubowchik GM, Conway CM, Coric V and Goadsby PJ: Rimegepant, an oral calcitonin gene-related peptide receptor antagonist, for migraine. N Engl J Med. 381:142–149. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Croop R, Goadsby PJ, Stock DA, Conway CM, Forshaw M, Stock EG, Coric V and Lipton RB: Efficacy, safety, and tolerability of rimegepant orally disintegrating tablet for the acute treatment of migraine: A randomised, phase 3, double-blind, placebo-controlled trial. Lancet. 394:737–745. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Portenoy RK, Payne D and Jacobsen P: Breakthrough pain: Characteristics and impact in patients with cancer pain. Pain. 81:129–134. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Truscott BM: Carcinoma of the breast; an analysis of the symptoms, factors affecting prognosis, results of treatment and recurrences in 1211 cases treated at the middlesex hospital. Br J Cancer. 1:129–145. 1947. View Article : Google Scholar : PubMed/NCBI

32 

Slagelse C, Munch T, Glazer C, Greene K, Finnerup NB, Kashani-Sabet M, Leong SP, Petersen KL and Rowbotham MC: Natural history of pain associated with melanoma surgery. Pain Rep. 3:e6892018. View Article : Google Scholar : PubMed/NCBI

33 

Yeh CF, Li WY, Chu PY, Kao SY, Chen YW, Lee TL, Hsu YB, Yang CC and Tai SK: Pretreatment pain predicts perineural invasion in oral squamous cell carcinoma: A prospective study. Oral Oncol. 61:115–119. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Brain SD: Sensory neuropeptides: Their role in inflammation and wound healing. Immunopharmacology. 37:133–152. 1997. View Article : Google Scholar : PubMed/NCBI

35 

Abdelaziz DM, Stone LS and Komarova SV: Localized experimental bone metastasis drives osteolysis and sensory hypersensitivity at distant non-tumor-bearing sites. Breast Cancer Res Treat. 153:9–20. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Auer J, Reeh PW and Fischer MJ: Acid-induced CGRP release from the stomach does not depend on TRPV1 or ASIC3. Neurogastroenterol Motil. 22:680–687. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Afroz S, Arakaki R, Iwasa T, Oshima M, Hosoki M, Inoue M, Baba O, Okayama Y and Matsuka Y: CGRP induces differential regulation of cytokines from satellite glial cells in trigeminal ganglia and orofacial nociception. Int J Mol Sci. 20:7112019. View Article : Google Scholar

38 

Hansen RR, Vacca V, Pitcher T, Clark AK and Malcangio M: Role of extracellular calcitonin gene-related peptide in spinal cord mechanisms of cancer-induced bone pain. Pain. 157:666–676. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Niiyama Y, Kawamata T, Yamamoto J, Omote K and Namiki A: Bone cancer increases transient receptor potential vanilloid subfamily 1 expression within distinct subpopulations of dorsal root ganglion neurons. Neuroscience. 148:560–572. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Jimenez-Andrade JM, Bloom AP, Stake JI, Mantyh WG, Taylor RN, Freeman KT, Ghilardi JR, Kuskowski MA and Mantyh PW: Pathological sprouting of adult nociceptors in chronic prostate cancer-induced bone pain. J Neurosci. 30:14649–14656. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Bloom AP, Jimenez-Andrade JM, Taylor RN, Castañeda-Corral G, Kaczmarska MJ, Freeman KT, Coughlin KA, Ghilardi JR, Kuskowski MA and Mantyh PW: Breast cancer-induced bone remodeling, skeletal pain, and sprouting of sensory nerve fibers. J Pain. 12:698–711. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Wakabayashi H, Wakisaka S, Hiraga T, Hata K, Nishimura R, Tominaga M and Yoneda T: Decreased sensory nerve excitation and bone pain associated with mouse Lewis lung cancer in TRPV1-deficient mice. J Bone Miner Metab. 36:274–285. 2018. View Article : Google Scholar : PubMed/NCBI

43 

Wacnik PW, Baker CM, Herron MJ, Kren BT, Blazar BR, Wilcox GL, Hordinsky MK, Beitz AJ and Ericson ME: Tumor-induced mechanical hyperalgesia involves CGRP receptors and altered innervation and vascularization of DsRed2 fluorescent hindpaw tumors. Pain. 115:95–106. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Nagamine K, Ozaki N, Shinoda M, Asai H, Nishiguchi H, Mitsudo K, Tohnai I, Ueda M and Sugiura Y: Mechanical allodynia and thermal hyperalgesia induced by experimental squamous cell carcinoma of the lower gingiva in rats. J Pain. 7:659–670. 2006. View Article : Google Scholar : PubMed/NCBI

45 

Levy MJ, Classey JD, Maneesri S, Meeran K, Powell M and Goadsby PJ: The association between calcitonin gene-related peptide (CGRP), substance P and headache in pituitary tumours. Pituitary. 7:67–71. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Dottorini ME, Assi A, Sironi M, Sangalli G, Spreafico G and Colombo L: Multivariate analysis of patients with medullary thyroid carcinoma. Prognostic significance and impact on treatment of clinical and pathologic variables. Cancer. 77:1556–1565. 1996. View Article : Google Scholar : PubMed/NCBI

47 

Goadsby PJ, Edvinsson L and Ekman R: Release of vasoactive peptides in the extracerebral circulation of humans and the cat during activation of the trigeminovascular system. Ann Neurol. 23:193–196. 1988. View Article : Google Scholar : PubMed/NCBI

48 

Kelley MJ, Snider RH, Becker KL and Johnson BE: Small cell lung carcinoma cell lines express mRNA for calcitonin and alpha- and beta-calcitonin gene related peptides. Cancer Lett. 81:19–25. 1994. View Article : Google Scholar : PubMed/NCBI

49 

Takami H, Shikata J, Horie H, Horiuchi J, Sakurai H and Ito K: Radioimmunoassay of plasma calcitonin gene-related peptide (CGRP) levels in patients with endocrine tumor. Gan To Kagaku Ryoho. 16:2219–2225. 1989.PubMed/NCBI

50 

Nagakawa O, Ogasawara M, Fujii H, Murakami K, Murata J, Fuse H and Saiki I: Effect of prostatic neuropeptides on invasion and migration of PC-3 prostate cancer cells. Cancer Lett. 133:27–33. 1998. View Article : Google Scholar : PubMed/NCBI

51 

Nagakawa O, Ogasawara M, Murata J, Fuse H and Saiki I: Effect of prostatic neuropeptides on migration of prostate cancer cell lines. Int J Urol. 8:65–70. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Wimalawansa SJ: CGRP radioreceptor assay: A new diagnostic tool for medullary thyroid carcinoma. J Bone Miner Res. 8:467–473. 1993. View Article : Google Scholar : PubMed/NCBI

53 

Angenendt L, Bormann E, Pabst C, Alla V, Görlich D, Braun L, Dohlich K, Schwöppe C, Bohlander SK, Arteaga MF, et al: The neuropeptide receptor calcitonin receptor-like (CALCRL) is a potential therapeutic target in acute myeloid leukemia. Leukemia. 33:2830–2841. 2019. View Article : Google Scholar : PubMed/NCBI

54 

Toda M, Suzuki T, Hosono K, Hayashi I, Hashiba S, Onuma Y, Amano H, Kurihara Y, Kurihara H, Okamoto H, et al: Neuronal system-dependent facilitation of tumor angiogenesis and tumor growth by calcitonin gene-related peptide. Proc Natl Acad Sci USA. 105:13550–13555. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Ostrovskaya A, Hick C, Hutchinson DS, Stringer BW, Wookey PJ, Wootten D, Sexton PM and Furness SGB: Expression and activity of the calcitonin receptor family in a sample of primary human high-grade gliomas. BMC Cancer. 19:1572019. View Article : Google Scholar : PubMed/NCBI

56 

Dallmayer M, Li J, Ohmura S, Alba Rubio R, Baldauf MC, Hölting TLB, Musa J, Knott MML, Stein S, Cidre-Aranaz F, et al: Targeting the CALCB/RAMP1 axis inhibits growth of Ewing sarcoma. Cell Death Dis. 10:1162019. View Article : Google Scholar : PubMed/NCBI

57 

Höppener JW, Steenbergh PH, Slebos RJ, Visser A, Lips CJ, Jansz HS, Bechet JM, Lenoir GM, Born W, Haller-Brem S, et al: Expression of the second calcitonin/calcitonin gene-related peptide gene in Ewing sarcoma cell lines. J Clin Endocrinol Metab. 64:809–817. 1987. View Article : Google Scholar : PubMed/NCBI

58 

Gutierrez S and Boada MD: Neuropeptide-induced modulation of carcinogenesis in a metastatic breast cancer cell line (MDA-MB-231LUC+). Cancer Cell Int. 18:2162018. View Article : Google Scholar : PubMed/NCBI

59 

Fristad I, Vandevska-Radunovic V, Fjeld K, Wimalawansa SJ and Hals Kvinnsland I: NK1, NK2, NK3 and CGRP1 receptors identified in rat oral soft tissues, and in bone and dental hard tissue cells. Cell Tissue Res. 311:383–391. 2003. View Article : Google Scholar : PubMed/NCBI

60 

Warburg O: On the origin of cancer cells. Science. 123:309–314. 1956. View Article : Google Scholar : PubMed/NCBI

61 

Sasahira T and Kirita T: Hallmarks of cancer-related newly prognostic factors of oral squamous cell carcinoma. Int J Mol Sci. 19:24132018. View Article : Google Scholar

62 

Kurihara-Shimomura M, Sasahira T, Nakashima C, Kuniyasu H, Shimomura H and Kirita T: The multifarious functions of pyruvate kinase M2 in oral cancer cells. Int J Mol Sci. 19:29072018. View Article : Google Scholar

63 

Lu H, Li X, Luo Z, Liu J and Fan Z: Cetuximab reverses the Warburg effect by inhibiting HIF-1-regulated LDH-A. Mol Cancer Ther. 12:2187–2199. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Zhai X, Yang Y, Wan J, Zhu R and Wu Y: Inhibition of LDH-A by oxamate induces G2/M arrest, apoptosis and increases radiosensitivity in nasopharyngeal carcinoma cells. Oncol Rep. 30:2983–2991. 2013. View Article : Google Scholar : PubMed/NCBI

65 

Rossetti L, Farrace S, Choi SB, Giaccari A, Sloan L, Frontoni S and Katz MS: Multiple metabolic effects of CGRP in conscious rats: Role of glycogen synthase and phosphorylase. Am J Physiol. 264:E1–E10. 1993.PubMed/NCBI

66 

Tang W and Fan Y: SIRT6 as a potential target for treating insulin resistance. Life Sci. 231:1165582019. View Article : Google Scholar : PubMed/NCBI

67 

Danaher RN, Loomes KM, Leonard BL, Whiting L, Hay DL, Xu LY, Kraegen EW, Phillips AR and Cooper GJ: Evidence that alpha-calcitonin gene-related peptide is a neurohormone that controls systemic lipid availability and utilization. Endocrinology. 149:154–160. 2008. View Article : Google Scholar : PubMed/NCBI

68 

Faubert B, Vincent EE, Poffenberger MC and Jones RG: The AMP-activated protein kinase (AMPK) and cancer: Many faces of a metabolic regulator. Cancer Lett. 356:165–170. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Zelissen PM, Koppeschaar HP, Lips CJ and Hackeng WH: Calcitonin gene-related peptide in human obesity. Peptides. 12:861–863. 1991. View Article : Google Scholar : PubMed/NCBI

70 

Gram DX, Hansen AJ, Wilken M, Elm T, Svendsen O, Carr RD, Ahrén B and Brand CL: Plasma calcitonin gene-related peptide is increased prior to obesity, and sensory nerve desensitization by capsaicin improves oral glucose tolerance in obese Zucker rats. Eur J Endocrinol. 153:963–969. 2005. View Article : Google Scholar : PubMed/NCBI

71 

Jan CI, Tsai MH, Chiu CF, Huang YP, Liu CJ and Chang NW: Fenofibrate suppresses oral tumorigenesis via reprogramming metabolic processes: Potential drug repurposing for oral cancer. Int J Biol Sci. 12:786–798. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Hu Q, Peng J, Chen X, Li H, Song M, Cheng B and Wu T: Obesity and genes related to lipid metabolism predict poor survival in oral squamous cell carcinoma. Oral Oncol. 89:14–22. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Eichel K, Jullié D and von Zastrow M: β-Arrestin drives MAP kinase signalling from clathrin-coated structures after GPCR dissociation. Nat Cell Biol. 18:303–310. 2016. View Article : Google Scholar : PubMed/NCBI

74 

Calebiro D, Nikolaev VO, Gagliani MC, de Filippis T, Dees C, Tacchetti C, Persani L and Lohse MJ: Persistent cAMP-signals triggered by internalized G-protein-coupled receptors. PLoS Biol. 7:e10001722009. View Article : Google Scholar : PubMed/NCBI

75 

Kuwasako K, Shimekake Y, Masuda M, Nakahara K, Yoshida T, Kitaura M, Kitamura K, Eto T and Sakata T: Visualization of the calcitonin receptor-like receptor and its receptor activity-modifying proteins during internalization and recycling. J Biol Chem. 275:29602–29609. 2000. View Article : Google Scholar : PubMed/NCBI

76 

Yarwood RE, Imlach WL, Lieu T, Veldhuis NA, Jensen DD, Klein Herenbrink C, Aurelio L, Cai Z, Christie MJ, Poole DP, et al: Endosomal signaling of the receptor for calcitonin gene-related peptide mediates pain transmission. Proc Natl Acad Sci USA. 114:12309–12314. 2017. View Article : Google Scholar : PubMed/NCBI

77 

Mantyh PW, DeMaster E, Malhotra A, Ghilardi JR, Rogers SD, Mantyh CR, Liu H, Basbaum AI, Vigna SR, Maggio JE, et al: Receptor endocytosis and dendrite reshaping in spinal neurons after somatosensory stimulation. Science. 268:1629–1632. 1995. View Article : Google Scholar : PubMed/NCBI

78 

Steinhoff MS, von Mentzer B, Geppetti P, Pothoulakis C and Bunnett NW: Tachykinins and their receptors: Contributions to physiological control and the mechanisms of disease. Physiol Rev. 94:265–301. 2014. View Article : Google Scholar : PubMed/NCBI

79 

Godbole A, Lyga S, Lohse MJ and Calebiro D: Publisher correction: Internalized TSH receptors en route to the TGN induce local Gs-protein signaling and gene transcription. Nat Commun. 9:54592018. View Article : Google Scholar : PubMed/NCBI

80 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

81 

Steinman L: Elaborate interactions between the immune and nervous systems. Nat Immunol. 5:575–581. 2004. View Article : Google Scholar : PubMed/NCBI

82 

Sternberg EM: Neural regulation of innate immunity: A coordinated nonspecific host response to pathogens. Nat Rev Immunol. 6:318–328. 2006. View Article : Google Scholar : PubMed/NCBI

83 

Antúnez C, Torres MJ, López S, Rodriguez-Pena R, Blanca M, Mayorga C and Santamaría-Babi LF: Calcitonin gene-related peptide modulates interleukin-13 in circulating cutaneous lymphocyte-associated antigen-positive T cells in patients with atopic dermatitis. Br J Dermatol. 161:547–553. 2009. View Article : Google Scholar : PubMed/NCBI

84 

Xing L, Guo J and Wang X: Induction and expression of beta-calcitonin gene-related peptide in rat T lymphocytes and its significance. J Immunol. 165:4359–4366. 2000. View Article : Google Scholar : PubMed/NCBI

85 

Bracci-Laudiero L, Aloe L, Buanne P, Finn A, Stenfors C, Vigneti E, Theodorsson E and Lundeberg T: NGF modulates CGRP synthesis in human B-lymphocytes: A possible anti-inflammatory action of NGF? J Neuroimmunol. 123:58–65. 2002. View Article : Google Scholar : PubMed/NCBI

86 

Bracci-Laudiero L, Aloe L, Caroleo MC, Buanne P, Costa N, Starace G and Lundeberg T: Endogenous NGF regulates CGRP expression in human monocytes, and affects HLA-DR and CD86 expression and IL-10 production. Blood. 106:3507–3514. 2005. View Article : Google Scholar : PubMed/NCBI

87 

Ding W, Stohl LL, Wagner JA and Granstein RD: Calcitonin gene-related peptide biases Langerhans cells toward Th2-type immunity. J Immunol. 181:6020–6026. 2008. View Article : Google Scholar : PubMed/NCBI

88 

Harzenetter MD, Novotny AR, Gais P, Molina CA, Altmayr F and Holzmann B: Negative regulation of TLR responses by the neuropeptide CGRP is mediated by the transcriptional repressor ICER. J Immunol. 179:607–615. 2007. View Article : Google Scholar : PubMed/NCBI

89 

Wang F, Millet I, Bottomly K and Vignery A: Calcitonin gene-related peptide inhibits interleukin 2 production by murine T lymphocytes. J Biol Chem. 267:21052–21057. 1992.PubMed/NCBI

90 

Kawamura N, Tamura H, Obana S, Wenner M, Ishikawa T, Nakata A and Yamamoto H: Differential effects of neuropeptides on cytokine production by mouse helper T cell subsets. Neuroimmunomodulation. 5:9–15. 1998. View Article : Google Scholar : PubMed/NCBI

91 

Hosoi J, Murphy GF, Egan CL, Lerner EA, Grabbe S, Asahina A and Granstein RD: Regulation of langerhans cell function by nerves containing calcitonin gene-related peptide. Nature. 363:159–163. 1993. View Article : Google Scholar : PubMed/NCBI

92 

Carucci JA, Ignatius R, Wei Y, Cypess AM, Schaer DA, Pope M, Steinman RM and Mojsov S: Calcitonin gene-related peptide decreases expression of HLA-DR and CD86 by human dendritic cells and dampens dendritic cell-driven T cell-proliferative responses via the type I calcitonin gene-related peptide receptor. J Immunol. 164:3494–3499. 2000. View Article : Google Scholar : PubMed/NCBI

93 

Holzmann B: Modulation of immune responses by the neuropeptide CGRP. Amino Acids. 45:1–7. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Nguyen MQ, Wu Y, Bonilla LS, von Buchholtz LJ and Ryba NJP: Diversity amongst trigeminal neurons revealed by high throughput single cell sequencing. PLoS One. 12:e01855432017. View Article : Google Scholar : PubMed/NCBI

95 

Zeisel A, Hochgerner H, Lönnerberg P, Johnsson A, Memic F, vander Zwan J, Häring M, Braun E, Borm LE, La Manno G, et al: Molecular architecture of the mouse nervous system. Cell. 174:999–1014 e22. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2020
Volume 20 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Lin C, Wang X and Ji T: Calcitonin gene‑related peptide: A promising bridge between cancer development and cancer‑associated pain in oral squamous cell carcinoma (Review). Oncol Lett 20: 253, 2020
APA
Zhang, Y., Lin, C., Wang, X., & Ji, T. (2020). Calcitonin gene‑related peptide: A promising bridge between cancer development and cancer‑associated pain in oral squamous cell carcinoma (Review). Oncology Letters, 20, 253. https://doi.org/10.3892/ol.2020.12116
MLA
Zhang, Y., Lin, C., Wang, X., Ji, T."Calcitonin gene‑related peptide: A promising bridge between cancer development and cancer‑associated pain in oral squamous cell carcinoma (Review)". Oncology Letters 20.5 (2020): 253.
Chicago
Zhang, Y., Lin, C., Wang, X., Ji, T."Calcitonin gene‑related peptide: A promising bridge between cancer development and cancer‑associated pain in oral squamous cell carcinoma (Review)". Oncology Letters 20, no. 5 (2020): 253. https://doi.org/10.3892/ol.2020.12116