Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Immunogenic camptothesome nanovesicles comprising sphingomyelin-derived camptothecin bilayers for safe and synergistic cancer immunochemotherapy

Abstract

Despite the enormous therapeutic potential of immune checkpoint blockade (ICB), it benefits only a small subset of patients. Some chemotherapeutics can switch ‘immune-cold’ tumours to ‘immune-hot’ to synergize with ICB. However, safe and universal therapeutic platforms implementing such immune effects remain scarce. We demonstrate that sphingomyelin-derived camptothecin nanovesicles (camptothesomes) elicit potent granzyme-B- and perforin-mediated cytotoxic T lymphocyte (CTL) responses, potentiating PD-L1/PD-1 co-blockade to eradicate subcutaneous MC38 adenocarcinoma with developed memory immunity. In addition, camptothesomes improve the pharmacokinetics and lactone stability of camptothecin, avoid systemic toxicities, penetrate deeply into the tumour and outperform the antitumour efficacy of Onivyde. Camptothesome co-load the indoleamine 2,3-dioxygenase inhibitor indoximod into its interior using the lipid-bilayer-crossing capability of the immunogenic cell death inducer doxorubicin, eliminating clinically relevant advanced orthotopic CT26-Luc tumours and late-stage B16-F10-Luc2 melanoma, and achieving complete metastasis remission when combined with ICB and folate targeting. The sphingomyelin-derived nanotherapeutic platform and doxorubicin-enabled transmembrane transporting technology are generalizable to various therapeutics, paving the way for transformation of the cancer immunochemotherapy paradigm.

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

Access options

Buy this article

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

Fig. 1: Development of SM-derived CPT liposomal nanovesicles (camptothesomes).
Fig. 2: Camptothesomes increased the MTD of CPT without inducing systemic toxicities in healthy mice.
Fig. 3: Improved circulation half-life and tumour delivery with efficient intratumoral drug release and deep tumour penetration.
Fig. 4: Camptothesome synergizes with PD-L1/PD-1 blockade to eradicate CRC tumours.
Fig. 5: Co-encapsulating DOX-IND in camptothesome-4 using DOX as a transmembrane-enabling carrier.
Fig. 6: Eradication of advanced and metastatic orthotopic CRC and subcutaneous melanoma tumours.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available within the article and the Supplementary Information. All relevant data are available from the authors upon reasonable request.

References

  1. Haslam, A. & Prasad, V. Estimation of the percentage of US patients with cancer who are eligible for and respond to checkpoint inhibitor immunotherapy drugs. JAMA Netw. Open 2, e192535 (2019).

    Article  Google Scholar 

  2. Catenacci, D. V. T., Hochster, H. & Klempner, S. J. Keeping checkpoint inhibitors in check. JAMA Netw. Open 2, e192546 (2019).

    Article  Google Scholar 

  3. Overman, M. J., Ernstoff, M. S. & Morse, M. A. Where we stand with immunotherapy in colorectal cancer: deficient mismatch repair, proficient mismatch repair, and toxicity management. Am. Soc. Clin. Oncol. Educ. Book 38, 239–247 (2018).

    Article  Google Scholar 

  4. Huyghe, N., Baldin, P. & Van den Eynde, M. Immunotherapy with immune checkpoint inhibitors in colorectal cancer: what is the future beyond deficient mismatch-repair tumours? Gastroenterol. Rep. 8, 11–24 (2020).

    Article  Google Scholar 

  5. Galon, J. & Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 18, 197–218 (2019).

    Article  CAS  Google Scholar 

  6. Song, W. et al. Synergistic and low adverse effect cancer immunotherapy by immunogenic chemotherapy and locally expressed PD-L1 trap. Nat. Commun. 9, 2237 (2018).

    Article  Google Scholar 

  7. Paz-Ares, L. et al. Pembrolizumab plus chemotherapy for squamous non-small-cell lung cancer. N. Engl. J. Med. 379, 2040–2051 (2018).

    Article  CAS  Google Scholar 

  8. Whiteside, T. L., Demaria, S., Rodriguez-Ruiz, M. E., Zarour, H. M. & Melero, I. Emerging opportunities and challenges in cancer immunotherapy. Clin. Cancer Res. 22, 1845–1855 (2016).

    Article  CAS  Google Scholar 

  9. Ebert, P. J. R. et al. MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade. Immunity 44, 609–621 (2016).

    Article  CAS  Google Scholar 

  10. Ribas, A. et al. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell 170, 1109–1119.e10 (2017).

    Article  CAS  Google Scholar 

  11. Chen, Y. F. et al. Host immune response to anti-cancer camptothecin conjugated cyclodextrin-based polymers. J. Biomed. Sci. 26, 85 (2019).

    Article  Google Scholar 

  12. Lu, J. et al. The self-assembling camptothecin-tocopherol prodrug: an effective approach for formulating camptothecin. Biomaterials 62, 176–187 (2015).

    Article  CAS  Google Scholar 

  13. Dosset, M. et al. PD-1/PD-L1 pathway: an adaptive immune resistance mechanism to immunogenic chemotherapy in colorectal cancer. Oncoimmunology 7, e1433981 (2018).

    Article  Google Scholar 

  14. Prendergast, G. C. et al. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol. Immunother. 63, 721–735 (2014).

    Article  CAS  Google Scholar 

  15. Hou, D. Y. et al. Inhibition of indoleamine 2,3-dioxygenase in dendritic cells by stereoisomers of 1-methyl-tryptophan correlates with antitumor responses. Cancer Res. 67, 792–801 (2007).

    Article  CAS  Google Scholar 

  16. Lob, S., Konigsrainer, A., Rammensee, H. G., Opelz, G. & Terness, P. Inhibitors of indoleamine-2,3-dioxygenase for cancer therapy: can we see the wood for the trees? Nat. Rev. Cancer 9, 445–452 (2009).

    Article  Google Scholar 

  17. Phan, T. et al. Salmonella-mediated therapy targeting indoleamine 2,3-dioxygenase 1 (IDO) activates innate immunity and mitigates colorectal cancer growth. Cancer Gene. Ther. 27, 235–245 (2020).

    Article  CAS  Google Scholar 

  18. Gomes, B. et al. Characterization of the selective indoleamine 2,3-dioxygenase-1 (IDO1) catalytic inhibitor EOS200271/PF-06840003 supports IDO1 as a critical resistance mechanism to PD-(L)1 blockade therapy. Mol. Cancer Ther. 17, 2530–2542 (2018).

    Article  CAS  Google Scholar 

  19. Metz, R. et al. IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: a novel IDO effector pathway targeted by D-1-methyl-tryptophan. Oncoimmunology 1, 1460–1468 (2012).

    Article  Google Scholar 

  20. Du, J. Z., Du, X. J., Mao, C. Q. & Wang, J. Tailor-made dual pH-sensitive polymer-doxorubicin nanoparticles for efficient anticancer drug delivery. J. Am. Chem. Soc. 133, 17560–17563 (2011).

    Article  CAS  Google Scholar 

  21. Muller, A. J., DuHadaway, J. B., Donover, P. S., Sutanto-Ward, E. & Prendergast, G. C. Inhibition of indoleamine 2,3-dioxygenase, an immunoregulatory target of the cancer suppression gene Bin1, potentiates cancer chemotherapy. Nat. Med. 11, 312–319 (2005).

    Article  CAS  Google Scholar 

  22. Kroemer, G., Galluzzi, L., Kepp, O. & Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 31, 51–72 (2013).

    Article  CAS  Google Scholar 

  23. Gamcsik, M. P., Kasibhatla, M. S., Teeter, S. D. & Colvin, O. M. Glutathione levels in human tumors. Biomarkers 17, 671–691 (2012).

    Article  CAS  Google Scholar 

  24. Zhong, Y. J., Shao, L. H. & Li, Y. Cathepsin B-cleavable doxorubicin prodrugs for targeted cancer therapy (Review). Int. J. Oncol. 42, 373–383 (2013).

    Article  CAS  Google Scholar 

  25. Shaw, J. P. & Chou, I. N. Elevation of intracellular glutathione content associated with mitogenic stimulation of quiescent fibroblasts. J. Cell. Physiol. 129, 193–198 (1986).

    Article  CAS  Google Scholar 

  26. Jessani, N., Liu, Y., Humphrey, M. & Cravatt, B. F. Enzyme activity profiles of the secreted and membrane proteome that depict cancer cell invasiveness. Proc. Natl Acad. Sci. USA 99, 10335–10340 (2002).

    Article  CAS  Google Scholar 

  27. Bucher, G. Ester pyrolysis of carbonates: bis(benzene hydrate) carbonate as potential precursor for monomeric carbonic acid. Eur. J. Org. Chem. 2010, 1070–1075 (2010).

    Article  Google Scholar 

  28. Bhattacharyya, J. et al. A paclitaxel-loaded recombinant polypeptide nanoparticle outperforms Abraxane in multiple murine cancer models. Nat. Commun. 6, 7939 (2015).

    Article  CAS  Google Scholar 

  29. Gabizon, A., Tzemach, D., Mak, L., Bronstein, M. & Horowitz, A. T. Dose dependency of pharmacokinetics and therapeutic efficacy of pegylated liposomal doxorubicin (DOXIL) in murine models. J. Drug Target 10, 539–548 (2002).

    Article  CAS  Google Scholar 

  30. Soundararajan, A., Bao, A., Phillips, W. T., Perez, R. 3rd & Goins, B. A. [(186)Re]liposomal doxorubicin (Doxil): in vitro stability, pharmacokinetics, imaging and biodistribution in a head and neck squamous cell carcinoma xenograft model. Nucl. Med. Biol. 36, 515–524 (2009).

    Article  CAS  Google Scholar 

  31. Kalra, A. et al. Abstract 2065: Magnetic resonance imaging with an iron oxide nanoparticle demonstrates the preclinical feasibility of predicting intratumoral uptake and activity of MM-398, a nanoliposomal irinotecan (nal-IRI). Cancer Res. 74, 2065–2065 (2014).

    Article  Google Scholar 

  32. Song, W. et al. Trapping of lipopolysaccharide to promote immunotherapy against colorectal cancer and attenuate liver metastasis. Adv. Mater. 30, e1805007 (2018).

    Article  Google Scholar 

  33. Low, P. S., Henne, W. A. & Doorneweerd, D. D. Discovery and development of folic-acid-based receptor targeting for imaging and therapy of cancer and inflammatory diseases. Acc. Chem. Res. 41, 120–129 (2008).

    Article  CAS  Google Scholar 

  34. Lu, J. et al. Targeted delivery of doxorubicin by folic acid-decorated dual functional nanocarrier. Mol. Pharmaceutics 11, 4164–4178 (2014).

    Article  CAS  Google Scholar 

  35. Zhang, Y., Davis, C., Ryan, J., Janney, C. & Pena, M. M. Development and characterization of a reliable mouse model of colorectal cancer metastasis to the liver. Clin. Exp. Metastasis 30, 903–918 (2013).

    Article  Google Scholar 

  36. Lu, J. et al. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat. Commun. 8, 1811 (2017).

    Article  Google Scholar 

  37. Slotte, J. P. The importance of hydrogen bonding in sphingomyelin’s membrane interactions with co-lipids. Biochim. Biophys. Acta 1858, 304–310 (2016).

    Article  CAS  Google Scholar 

  38. Narvekar, M., Xue, H. Y., Eoh, J. Y. & Wong, H. L. Nanocarrier for poorly water-soluble anticancer drugs—barriers of translation and solutions. AAPS PharmSciTech 15, 822–833 (2014).

    Article  CAS  Google Scholar 

  39. Lu, J. et al. PEG-derivatized embelin as a nanomicellar carrier for delivery of paclitaxel to breast and prostate cancers. Biomaterials 34, 1591–1600 (2013).

    Article  CAS  Google Scholar 

  40. Lu, J. et al. An improved d-alpha-tocopherol-based nanocarrier for targeted delivery of doxorubicin with reversal of multidrug resistance. J. Control. Release 196, 272–286 (2014).

    Article  CAS  Google Scholar 

  41. Rao, Y. et al. Bouchardatine analogue alleviates non-alcoholic hepatic fatty liver disease/non-alcoholic steatohepatitis in high-fat fed mice by inhibiting ATP synthase activity. Br. J. Pharmacol. 176, 2877–2893 (2019).

    Article  CAS  Google Scholar 

  42. Konstantopoulos, P. et al. Metabolic effects of Crocus sativus and protective action against non-alcoholic fatty liver disease in diabetic rats. Biomed. Rep. 6, 513–518 (2017).

    CAS  Google Scholar 

  43. Abdel-Salam, O. M. E., Nada, S. A., Salem, N. A., El-Shamarka, M. E.-S. & Omara, E. Effect of Cannabis sativa on oxidative stress and organ damage after systemic endotoxin administration in mice. Comp. Clin. Pathol. 23, 1069–1085 (2013).

    Article  Google Scholar 

  44. Clemente-Casares, X. et al. Expanding antigen-specific regulatory networks to treat autoimmunity. Nature 530, 434–440 (2016).

    Article  CAS  Google Scholar 

  45. Wang, W. et al. RIP1 kinase drives macrophage-mediated adaptive immune tolerance in pancreatic cancer. Cancer Cell 34, 757–774.e7 (2018).

    Article  CAS  Google Scholar 

  46. Zhang, Y., Huo, M., Zhou, J. & Xie, S. PKSolver: an add-in program for pharmacokinetic and pharmacodynamic data analysis in Microsoft Excel. Comput. Meth. Prog. Bio. 99, 306–314 (2010).

    Article  Google Scholar 

  47. Litzenburger, U. M. et al. Constitutive IDO expression in human cancer is sustained by an autocrine signaling loop involving IL-6, STAT3 and the AHR. Oncotarget 5, 1038–1051 (2014).

    Article  Google Scholar 

Download references

Acknowledgements

This work was supported in part by a Startup Fund from the College of Pharmacy at the University of Arizona and two Seed Grants from the University of Arizona BIO5 Institute and the State of Arizona’s Technology and Research Initiative Fund (TRIF), and by National Institutes of Health (NIH) grants (NIEHS P30 ES006694, NCI P30 CA023074 and NCI R01 CA092596). We acknowledge Statistical Consulting at the University of Arizona Information Technology for their assistance with the statistical analysis; the use of mass spectrometry equipment in the Analytical and Biological Mass Spectrometry Core Facility at the University of Arizona BIO5 Institute; the University of Arizona Translational Bioimaging Resource Core for the Lago live animal imaging; the University of Arizona University Animal Care Pathology Services for the serum chemistry and haematological counts; the TACMASR of the UACC for the IHC and H&E staining, immunofluorescence and confocal laser scanning microscopy; and Arizona State University’s John Cowley Center for High Resolution Electron Microscopy (the specific instrumentation used was supported by the National Science Foundation (NSF), MRI grant NSF1531991) for the cryo-EM.

Author information

Authors and Affiliations

Authors

Contributions

J.L. conceived and supervised the project. J.L. and Z.W. designed the experiments, analysed the data and wrote the manuscript. Z.W. performed the experiments. N.L., J.C., K.T.L. and K.T.L. assisted in the prodrug synthesis, nanoparticle preparation, HPLC and in vivo animal studies. W.H. assisted with the PCR assay. A.J.S. provided clinical research insight into antitumour efficacy studies. All of the authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Jianqin Lu.

Ethics declarations

Competing interests

J.L. has applied for patents related to this study. The other authors have no competing interests.

Additional information

Peer review information Nature Nanotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Materials, Methods, Figs. 1–35 and refs. 1–7.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Z., Little, N., Chen, J. et al. Immunogenic camptothesome nanovesicles comprising sphingomyelin-derived camptothecin bilayers for safe and synergistic cancer immunochemotherapy. Nat. Nanotechnol. 16, 1130–1140 (2021). https://doi.org/10.1038/s41565-021-00950-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41565-021-00950-z

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research