nature.com

Nociceptor neurons promote Pdac progression and cancer pain by interaction with cancer-associated fibroblasts and…

Abstract

The emerging field of cancer neuroscience has demonstrated great progress in revealing the crucial role of the nervous system in cancer initiation and progression. Pancreatic ductal adenocarcinoma (PDAC) is characterized by perineural invasion and modulated by autonomic (sympathetic and parasympathetic) and sensory innervations. Here, we further demonstrated that within the tumor microenvironment of PDAC, nociceptor neurons interacted with cancer-associated fibroblasts (CAFs) through calcitonin gene-related peptide (CGRP) and nerve growth factor (NGF). This interaction led to the inhibition of interleukin-15 expression in CAFs, suppressing the infiltration and cytotoxic function of natural killer (NK) cells and thereby promoting PDAC progression and cancer pain. In PDAC patients, nociceptive innervation of tumor tissue is negatively correlated with the infiltration of NK cells while positively correlated with pain intensity. This association serves as an independent prognostic factor for both overall survival and relapse-free survival for PDAC patients. Our findings highlight the crucial regulation of NK cells by nociceptor neurons through interaction with CAFs in the development of PDAC. We also propose that targeting nociceptor neurons or CGRP signaling may offer a promising therapy for PDAC and cancer pain.

This is a preview of subscription content, access via your institution

Access options

Access through your institution

Change institution

Buy or subscribe

Subscribe to this journal

Receive 12 digital issues and online access to articles

$119.00 per year

only $9.92 per issue

Learn more

Buy this article

Purchase on SpringerLink

Instant access to full article PDF

Buy now

Prices may be subject to local taxes which are calculated during checkout

Additional access options:

Log in

Learn about institutional subscriptions

Read our FAQs

Contact customer support

Fig. 1: Ablation of nociceptor neurons suppresses PDAC development and cancer pain.

Fig. 2: Ablation of nociceptor neurons improves the infiltration and cytotoxic function of NK cells.

Fig. 3: NK cells but not T cells mediated the inhibitory effect of nociceptor neurons on PDAC.

Fig. 4: CGRP modulates NK cells contributing to PDAC progression.

Fig. 5: IL-15 mediates the modulation of CGRP on NK cells.

Fig. 6: CGPR inhibits IL-15 secretion from CAFs.

Fig. 7: Interaction of nociceptive neurons with CAFs to suppress NK cells.

Fig. 8: Negative correlation of nociceptive innervation with survival of PDAC patients.

Data availability

Deidentified scRNA-seq raw data are available from the National Genomics Data Center (https://ngdc.cncb.ac.cn/subcenter/1) under accession number OEP005530 (Shared URL: https://www.biosino.org/node/project/detail/OEP005530). Further information related to the data reported in this paper can be acquired from the lead contact Jihui Hao (haojihui@tjmuch.com) upon reasonable request.

References

Monje, M. et al. Roadmap for the emerging field of cancer neuroscience. Cell 181, 219–222 (2020).

CASPubMedPubMed CentralGoogle Scholar

Winkler, F. et al. Cancer neuroscience: state of the field, emerging directions. Cell 186, 1689–1707 (2023).

CASPubMedPubMed CentralGoogle Scholar

Mancusi, R. & Monje, M. The neuroscience of cancer. Nature 618, 467–479 (2023).

CASPubMedPubMed CentralGoogle Scholar

Wang, K. et al. STING suppresses bone cancer pain via immune and neuronal modulation. Nat. Commun. 12, 4558 (2021).

CASPubMedPubMed CentralGoogle Scholar

Hanahan, D. & Monje, M. Cancer hallmarks intersect with neuroscience in the tumor microenvironment. Cancer Cell 41, 573–580 (2023).

CASPubMedPubMed CentralGoogle Scholar

Shi, D. D. et al. Therapeutic avenues for cancer neuroscience: translational frontiers and clinical opportunities. Lancet Oncol. 23, e62–e74 (2022).

CASPubMedPubMed CentralGoogle Scholar

Ferdoushi, A. et al. Tumor innervation and clinical outcome in pancreatic cancer. Sci. Rep. 11, 7390 (2021).

CASPubMedPubMed CentralGoogle Scholar

Kondo, N. et al. An increased number of perineural invasions is independently associated with poor survival of patients with resectable pancreatic ductal adenocarcinoma. Pancreas 44, 1345–1351 (2015).

PubMedGoogle Scholar

Crippa, S. et al. Implications of perineural invasion on disease recurrence and survival after pancreatectomy for pancreatic head ductal adenocarcinoma. Ann. Surg. 276, 378–385 (2022).

PubMedGoogle Scholar

Hinshaw, D. C. & Shevde, L. A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 79, 4557–4566 (2019).

CASPubMedPubMed CentralGoogle Scholar

Wang, H. et al. Role of the nervous system in cancers: a review. Cell Death Discov. 7, 76 (2021).

PubMedPubMed CentralGoogle Scholar

Demir, I. E. et al. Targeting nNOS ameliorates the severe neuropathic pain due to chronic pancreatitis. EBioMedicine 46, 431–443 (2019).

CASPubMedPubMed CentralGoogle Scholar

Ceyhan, G. O. et al. Pancreatic neuropathy results in “neural remodeling” and altered pancreatic innervation in chronic pancreatitis and pancreatic cancer. Am. J. Gastroenterol. 104, 2555–2565 (2009).

PubMedGoogle Scholar

Grossberg, A. J. et al. Multidisciplinary standards of care and recent progress in pancreatic ductal adenocarcinoma. CA Cancer J. Clin. 70, 375–403 (2020).

PubMedPubMed CentralGoogle Scholar

Ni, B. et al. Crosstalk between peripheral innervation and pancreatic ductal adenocarcinoma. Neurosci. Bull. 39, 1717–1731 (2023).

CASPubMedPubMed CentralGoogle Scholar

Saloman, J. L. et al. Ablation of sensory neurons in a genetic model of pancreatic ductal adenocarcinoma slows initiation and progression of cancer. Proc. Natl. Acad. Sci. USA 113, 3078–3083 (2016).

CASPubMedPubMed CentralGoogle Scholar

Sinha, S. et al. PanIN neuroendocrine cells promote tumorigenesis via neuronal cross-talk. Cancer Res. 77, 1868–1879 (2017).

CASPubMedPubMed CentralGoogle Scholar

Wu, M. et al. Innervation of nociceptor neurons in the spleen promotes germinal center responses and humoral immunity. Cell 187, 2935–2951.e19 (2024).

CASPubMedGoogle Scholar

Balood, M. et al. Nociceptor neurons affect cancer immunosurveillance. Nature 611, 405–412 (2022).

CASPubMedPubMed CentralGoogle Scholar

Pinho-Ribeiro, F. A. et al. Blocking neuronal signaling to immune cells treats streptococcal invasive infection. Cell 173, 1083–1097.e22 (2018).

CASPubMedPubMed CentralGoogle Scholar

Baral, P. et al. Nociceptor sensory neurons suppress neutrophil and γδ T cell responses in bacterial lung infections and lethal pneumonia. Nat. Med. 24, 417–426 (2018).

CASPubMedPubMed CentralGoogle Scholar

Tamari, M. et al. Sensory neurons promote immune homeostasis in the lung. Cell 187, 44–61.e17 (2024).

CASPubMedGoogle Scholar

Ceyhan, G. O. et al. Pancreatic neuropathy and neuropathic pain—a comprehensive pathomorphological study of 546 cases. Gastroenterology 136, 177–186.e1 (2009).

PubMedGoogle Scholar

Selvaraj, D., Hirth, M., Gandla, J. & Kuner, R. A mouse model for pain and neuroplastic changes associated with pancreatic ductal adenocarcinoma. Pain 158, 1609–1621 (2017).

PubMedGoogle Scholar

Li, L. et al. The impact of TRPV1 on cancer pathogenesis and therapy: a systematic review. Int J. Biol. Sci. 17, 2034–2049 (2021).

CASPubMedPubMed CentralGoogle Scholar

Lai, N. Y. et al. Gut-innervating nociceptor neurons regulate Peyer’s Patch Microfold cells and SFB levels to mediate Salmonella host defense. Cell 180, 33–49.e22 (2020).

CASPubMedGoogle Scholar

Wang, X. et al. Phenotype screens of murine pancreatic cancer identify a Tgf-α-Ccl2-paxillin axis driving human-like neural invasion. J. Clin. Invest. 133, e166333 (2023).

CASPubMedPubMed CentralGoogle Scholar

Makhmutova, M. & Caicedo, A. Optical imaging of pancreatic innervation. Front. Endocrinol. 12, 663022 (2021).

Google Scholar

Schwartz, E. S. et al. TRPV1 and TRPA1 antagonists prevent the transition of acute to chronic inflammation and pain in chronic pancreatitis. J. Neurosci. 33, 5603–5611 (2013).

CASPubMedPubMed CentralGoogle Scholar

Ma, Y. et al. Combination of PD-1 inhibitor and OX40 agonist induces tumor rejection and immune memory in mouse models of pancreatic cancer. Gastroenterology 159, 306–319.e12 (2020).

CASPubMedGoogle Scholar

Wang, K. et al. PD-1 blockade inhibits osteoclast formation and murine bone cancer pain. J. Clin. Invest. 130, 3603–3620 (2020).

CASPubMedPubMed CentralGoogle Scholar

Wakabayashi, H. et al. 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).

CASPubMedGoogle Scholar

Zhang, Y., Chen, M., Liu, Z., Wang, X. & Ji, T. The neuropeptide calcitonin gene-related peptide links perineural invasion with lymph node metastasis in oral squamous cell carcinoma. BMC Cancer 21, 1254 (2021).

CASPubMedPubMed CentralGoogle Scholar

Croop, R. et al. Oral rimegepant for preventive treatment of migraine: a phase 2/3, randomised, double-blind, placebo-controlled trial. Lancet 397, 51–60 (2021).

CASPubMedGoogle Scholar

Lipton, R. B. et al. Rimegepant, an oral calcitonin gene-related peptide receptor antagonist, for migraine. N. Engl. J. Med. 381, 142–149 (2019).

CASPubMedGoogle Scholar

Shimasaki, N., Jain, A. & Campana, D. NK cells for cancer immunotherapy. Nat. Rev. Drug Discov. 19, 200–218 (2020).

CASPubMedGoogle Scholar

Dean, I. et al. Rapid functional impairment of natural killer cells following tumor entry limits anti-tumor immunity. Nat. Commun. 15, 683 (2024).

CASPubMedPubMed CentralGoogle Scholar

Wang, D. & Wei, H. Natural killer cells in tumor immunotherapy. Cancer Biol. Med. 20, 539–544 (2023).

PubMedPubMed CentralGoogle Scholar

Kurz, E. et al. Exercise-induced engagement of the IL-15/IL-15Rα axis promotes anti-tumor immunity in pancreatic cancer. Cancer Cell 40, 720–737.e5 (2022).

CASPubMedPubMed CentralGoogle Scholar

Rebelo, R., Xavier, C. P. R., Giovannetti, E. & Vasconcelos, M. H. Fibroblasts in pancreatic cancer: molecular and clinical perspectives. Trends Mol. Med. 29, 439–453 (2023).

CASPubMedGoogle Scholar

Li, X. et al. Sonic hedgehog paracrine signaling activates stromal cells to promote perineural invasion in pancreatic cancer. Clin. Cancer Res. 20, 4326–4338 (2014).

CASPubMedGoogle Scholar

Stopczynski, R. E. et al. Neuroplastic changes occur early in the development of pancreatic ductal adenocarcinoma. Cancer Res. 74, 1718–1727 (2014).

CASPubMedPubMed CentralGoogle Scholar

Saloman, J. L. et al. Systemic depletion of nerve growth factor inhibits disease progression in a genetically engineered model of pancreatic ductal adenocarcinoma. Pancreas 47, 856–863 (2018).

CASPubMedPubMed CentralGoogle Scholar

Ye, Y., Xie, T. & Amit, M. Targeting the nerve-cancer circuit. Cancer Res. 83, 2445–2447 (2023).

CASPubMedGoogle Scholar

Wang, B. et al. Combinatorial sympathetic and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) blockades inhibit the murine melanoma growth by targeting infiltrating T cells. Transl. Cancer Res. 10, 899–913 (2021).

CASPubMedPubMed CentralGoogle Scholar

Globig, A. M. et al. The β(1)-adrenergic receptor links sympathetic nerves to T cell exhaustion. Nature 622, 383–392 (2023).

CASPubMedPubMed CentralGoogle Scholar

Yang, M. W. et al. Perineural invasion reprograms the immune microenvironment through cholinergic signaling in pancreatic ductal adenocarcinoma. Cancer Res. 80, 1991–2003 (2020).

CASPubMedGoogle Scholar

Szallasi, A. Resiniferatoxin: Nature’s precision medicine to silence TRPV1-positive afferents. Int. J. Mol. Sci. 24, 15042 (2023).

CASPubMedPubMed CentralGoogle Scholar

Brown, D. C. Resiniferatoxin: The evolution of the “Molecular Scalpel” for chronic pain relief. Pharmaceuticals 9, 47 (2016).

PubMedPubMed CentralGoogle Scholar

Brown, D. C., Agnello, K. & Iadarola, M. J. Intrathecal resiniferatoxin in a dog model: efficacy in bone cancer pain. Pain 156, 1018–1024 (2015).

CASPubMedPubMed CentralGoogle Scholar

Schnittert, J., Bansal, R. & Prakash, J. Targeting pancreatic stellate cells in cancer. Trends Cancer 5, 128–142 (2019).

CASPubMedGoogle Scholar

Sparmann, G. et al. Inhibition of lymphocyte apoptosis by pancreatic stellate cells: impact of interleukin-15. Am. J. Physiol. Gastrointest. Liver Physiol. 289, G842–G851 (2005).

CASPubMedGoogle Scholar

Ma, S., Caligiuri, M. A. & Yu, J. Harnessing IL-15 signaling to potentiate NK cell-mediated cancer immunotherapy. Trends Immunol. 43, 833–847 (2022).

CASPubMedPubMed CentralGoogle Scholar

Vivier, E. et al. Natural killer cell therapies. Nature 626, 727–736 (2024).

CASPubMedGoogle Scholar

Renz, B. W. et al. β2 Adrenergic-neurotrophin feedforward loop promotes pancreatic cancer. Cancer Cell 33, 75–90.e7 (2018).

CASPubMedGoogle Scholar

Ramer, M. S., Bradbury, E. J., Michael, G. J., Lever, I. J. & McMahon, S. B. Glial cell line-derived neurotrophic factor increases calcitonin gene-related peptide immunoreactivity in sensory and motoneurons in vivo. Eur. J. Neurosci. 18, 2713–2721 (2003).

PubMedGoogle Scholar

Kobayashi, H. et al. Neuro-mesenchymal interaction mediated by a β2 adrenergic-nerve growth factor feedforward loop promotes colorectal cancer progression. Cancer Discov. 15, 202–226 (2025).

PubMedGoogle Scholar

Bennett, M. I. Mechanism-based cancer-pain therapy. Pain 158, S74–S78 (2017).

PubMedGoogle Scholar

Schweizerhof, M. et al. Hematopoietic colony-stimulating factors mediate tumor-nerve interactions and bone cancer pain. Nat. Med. 15, 802–807 (2009).

CASPubMedGoogle Scholar

Selvaraj, D. et al. A functional role for VEGFR1 expressed in peripheral sensory neurons in cancer pain. Cancer Cell 27, 780–796 (2015).

CASPubMedPubMed CentralGoogle Scholar

Jimenez-Andrade, J. M., Ghilardi, J. R., Castañeda-Corral, G., Kuskowski, M. A. & Mantyh, P. W. Preventive or late administration of anti-NGF therapy attenuates tumor-induced nerve sprouting, neuroma formation, and cancer pain. Pain 152, 2564–2574 (2011).

CASPubMedPubMed CentralGoogle Scholar

Zhao, T. et al. ESE3-positive PSCs drive pancreatic cancer fibrosis, chemoresistance and poor prognosis via tumour-stromal IL-1β/NF-κB/ESE3 signalling axis. Br. J. Cancer 127, 1461–1472 (2022).

CASPubMedPubMed CentralGoogle Scholar

Liu, J. et al. Tumoral EHF predicts the efficacy of anti-PD1 therapy in pancreatic ductal adenocarcinoma. J. Exp. Med. 216, 656–673 (2019).

CASPubMedPubMed CentralGoogle Scholar

Donnelly, C. R. et al. STING controls nociception via type I interferon signalling in sensory neurons. Nature 591, 275–280 (2021).

CASPubMedPubMed CentralGoogle Scholar

Luo, X. et al. IL-23/IL-17A/TRPV1 axis produces mechanical pain via macrophage-sensory neuron crosstalk in female mice. Neuron 109, 2691–2706.e5 (2021).

CASPubMedPubMed CentralGoogle Scholar

Lovell, M. R. et al. Effect of cancer pain guideline implementation on pain outcomes among adult outpatients with cancer-related pain: A stepped wedge cluster randomized trial. JAMA Netw. Open 5, e220060 (2022).

PubMedPubMed CentralGoogle Scholar

de Conno, F. et al. Pain measurement in cancer patients: a comparison of six methods. Pain 57, 161–166 (1994).

PubMedGoogle Scholar

Gerbershagen, H. J., Rothaug, J., Kalkman, C. J. & Meissner, W. Determination of moderate-to-severe postoperative pain on the numeric rating scale: a cut-off point analysis applying four different methods. Br. J. Anaesth. 107, 619–626 (2011).

CASPubMedGoogle Scholar

Download references

Acknowledgements

The authors would thank Prof. Zilong Wang (Southern University of Science and Technology, China) for providing TRPV1-Cre and DTR mice. This work was funded by the National Natural Science Foundation of China (82030092 and 82273362), Major Project of Tianjin Public Health Science and Technology Program (24ZXGZSY00020), Tianjin Key Medical Discipline (Specialty) Construction Project (TJYXZDXK-009A), and State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine Project (QZ23-1).

Author information

Author notes

These authors contributed equally: Kaiyuan Wang, Bo Ni, Yongjie Xie.

Authors and Affiliations

Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China

Kaiyuan Wang & Limei Yuan

Pancreas Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Key Laboratory of Digestive Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin, China

Bo Ni, Yongjie Xie, Zekun Li, Chenyang Meng, Tiansuo Zhao, Song Gao, Chongbiao Huang, Hongwei Wang, Ying Ma, Tianxing Zhou, Yukuan Feng, Antao Chang, Chao Yang, Jun Yu, Wenwen Yu, Fenglin Zang, Yanhui Zhang, Xiuchao Wang & Jihui Hao

Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA

Ru-Rong Ji

Department of Cell Biology, Duke University Medical Center, Durham, NC, USA

Ru-Rong Ji

Department of Neurobiology, Duke University Medical Center, Durham, NC, USA

Ru-Rong Ji

Authors

Kaiyuan Wang

View author publications

You can also search for this author inPubMedGoogle Scholar

2. Bo Ni

View author publications

You can also search for this author inPubMedGoogle Scholar

3. Yongjie Xie

View author publications

You can also search for this author inPubMedGoogle Scholar

4. Zekun Li

View author publications

You can also search for this author inPubMedGoogle Scholar

5. Limei Yuan

View author publications

You can also search for this author inPubMedGoogle Scholar

6. Chenyang Meng

View author publications

You can also search for this author inPubMedGoogle Scholar

7. Tiansuo Zhao

View author publications

You can also search for this author inPubMedGoogle Scholar

8. Song Gao

View author publications

You can also search for this author inPubMedGoogle Scholar

9. Chongbiao Huang

View author publications

You can also search for this author inPubMedGoogle Scholar

10. Hongwei Wang

View author publications

You can also search for this author inPubMedGoogle Scholar

11. Ying Ma

View author publications

You can also search for this author inPubMedGoogle Scholar

12. Tianxing Zhou

View author publications

You can also search for this author inPubMedGoogle Scholar

13. Yukuan Feng

View author publications

You can also search for this author inPubMedGoogle Scholar

14. Antao Chang

View author publications

You can also search for this author inPubMedGoogle Scholar

15. Chao Yang

View author publications

You can also search for this author inPubMedGoogle Scholar

16. Jun Yu

View author publications

You can also search for this author inPubMedGoogle Scholar

17. Wenwen Yu

View author publications

You can also search for this author inPubMedGoogle Scholar

18. Fenglin Zang

View author publications

You can also search for this author inPubMedGoogle Scholar

19. Yanhui Zhang

View author publications

You can also search for this author inPubMedGoogle Scholar

20. Ru-Rong Ji

View author publications

You can also search for this author inPubMedGoogle Scholar

21. Xiuchao Wang

View author publications

You can also search for this author inPubMedGoogle Scholar

22. Jihui Hao

View author publications

You can also search for this author inPubMedGoogle Scholar

Contributions

J.H., R.R.J., X.W., and K.W. conceived and designed the study. K.W., B.N., Y.X., Z.L., L.Y., C.M., S.G., H.W., Y.M., T.X.Z., W.Y., F.Z., and Y.Z. performed the experiments. C.H., Y.X., T.S.Z., Y.F., A.C., C.Y., and J.Y. carried out data analysis. K.W. and X.W. wrote the first drafts of the manuscript; R.R.J. and J.H. edited the manuscript. Illustrations were created by B.N. and Y.X. using BioRender. All authors had full access to the data and approved the final version. J.H., X.W., R.R.J., and K.W. were responsible for the decision to submit the manuscript.

Corresponding authors

Correspondence to Kaiyuan Wang, Ru-Rong Ji, Xiuchao Wang or Jihui Hao.

Ethics declarations

Competing interests

The authors declare no competing interests.

Supplementary information

Supplementary information, Fig. S1

Supplementary information, Fig. S2

Supplementary information, Fig. S3

Supplementary information, Fig. S4

Supplementary information, Fig. S5

Supplementary information, Fig. S6

Supplementary information, Fig. S7

Supplementary information, Fig. S8

Supplementary information, Fig. S9

Supplementary information, Fig. S10

Supplementary information, Fig. S11

Supplementary information, Fig. S12

Supplementary information, Fig. S13

Supplementary information, Fig. S14

Supplementary information, Fig. S15

Supplementary information, Fig. S16

Supplementary information, Fig. S17

Supplementary information, Table S1

Supplementary information, Table S2

Supplementary information, Table S3

Supplementary information, Data S1

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, K., Ni, B., Xie, Y. et al. Nociceptor neurons promote PDAC progression and cancer pain by interaction with cancer-associated fibroblasts and suppression of natural killer cells. Cell Res (2025). https://doi.org/10.1038/s41422-025-01098-4

Download citation

Received:17 September 2024

Accepted:05 March 2025

Published:24 March 2025

DOI:https://doi.org/10.1038/s41422-025-01098-4

Share this article

Anyone you share the following link with will be able to read this content:

Get shareable link

Sorry, a shareable link is not currently available for this article.

Copy to clipboard

Provided by the Springer Nature SharedIt content-sharing initiative

Read full news in source page