We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

Immunotherapeutic effect of photothermal-mediated exosomes secreted from breast cancer cells

    Meysam Najaflou

    Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran

    Drug Applied Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran

    ,
    Farhad Bani

    Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran

    &
    Ahmad Yari Khosroushahi

    *Author for correspondence:

    E-mail Address: Yarikhosroushahia@tbzmed.ac.ir

    Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran

    Drug Applied Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran

    Published Online:https://doi.org/10.2217/nnm-2023-0014

    Aim: Exosomal damage-associated molecular patterns can play a key role in immunostimulation and changing the cold tumor microenvironment to hot. Materials & methods: This study examined the immunostimulation effect of photothermal and hyperthermia-treated 4T1 cell-derived exosomes on 4T1 cell-induced breast tumors in BALB/c animal models. Exosomes were characterized for HSP70, HSP90 and HMGB-1 before injection into mice and tumor tissues were analyzed for IL-6, IL-12 and IL-1β, CD4 and CD8 T-cell permeability, and PD-L1 expression. Results: Thermal treatments increased high damage-associated molecular patterns containing exosome secretion and the permeability of T cells to tumors, leading to tumor growth inhibition. Conclusion: Photothermal-derived exosomes showed higher damage-associated molecular patterns than hyperthermia with a higher immunostimulation and inhibiting tumor growth effect.

    Plain language summary

    This research explored the impact of using tiny dying cancer cell-derived particles known as exosomes to activate the immune system to fight against breast tumors in animal models. These exosomes contain specific molecules that can trigger the immune response and alter the environment surrounding the tumor. Researchers applied two different treatments, photothermal and hyperthermia, to kill the cancer cells and obtain these exosomes. Both treatments involved using heat to kill the cells. The study revealed that exosomes derived through the photothermal method exhibited higher levels of these immune-activating molecules compared with those obtained through hyperthermia. Upon injecting these exosomes into the animal models, they enhanced the ability of the immune cells to enter the tumors, resulting in a reduction in tumor growth. Overall, the findings indicate that using exosomes obtained through the photothermal method may be more effective in stimulating the immune system to fight against cancer and inhibiting tumor growth, as opposed to using exosomes obtained through hyperthermia.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Recht A, Come SE, Henderson IC et al. The sequencing of chemotherapy and radiation therapy after conservative surgery for early-stage breast cancer. N. Engl. J. Med. 334(21), 1356–1361 (1996).
    • 2. Mazzaferri EL, Jhiang SM. Long-term impact of initial surgical and medical therapy on papillary and follicular thyroid cancer. Am. J. Med. 97(5), 418–428 (1994).
    • 3. Wang X, Yang L, Chen Z, Shin DM. Application of nanotechnology in cancer therapy and imaging. CA Cancer J. Clin. 58(2), 97–110 (2008).
    • 4. Liu Y, Bhattarai P, Dai Z, Chen X. Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer. Chem. Soc. Rev. 48(7), 2053–2108 (2019). • Comprehensive review on different agents that can also be used in photothermal therapy and photoacoustic imaging that are promising next-generation noninvasive cancer theranostic techniques.
    • 5. Li C, Xu Y, Tu L et al. Rationally designed Ru (II)-metallacycle chemo-phototheranostic that emits beyond 1000 nm. Chem. Sci. 13(22), 6541–6549 (2022).
    • 6. Chen Y-W, Su Y-L, Hu S-H, Chen S-Y. Functionalized graphene nanocomposites for enhancing photothermal therapy in tumor treatment. Adv. Drug Deliv. Rev. 105, 190–204 (2016).
    • 7. Zhao L, Zhang X, Wang X, Guan X, Zhang W, Ma J. Recent advances in selective photothermal therapy of tumor. J. Nanobiotechnol. 19(1), 1–15 (2021).
    • 8. Huang X, Jain PK, El-Sayed IH, El-Sayed MA. Plasmonic photothermal therapy (PPTT) using gold nanoparticles. Lasers Med. Sci. 23(3), 217–228 (2008).
    • 9. Kuo WS, Chang CN, Chang YT et al. Gold nanorods in photodynamic therapy, as hyperthermia agents, and in near-infrared optical imaging. Angewandte Chemie 122(15), 2771–2775 (2010).
    • 10. Kim HS, Lee DY. Near-infrared-responsive cancer photothermal and photodynamic therapy using gold nanoparticles. Polymers 10(9), 961 (2018).
    • 11. Breitenborn H, Dong J, Piccoli R et al. Quantifying the photothermal conversion efficiency of plasmonic nanoparticles by means of terahertz radiation. APL Photon. 4(12), 1261061–1261069 (2019).
    • 12. Li X, Yong T, Wei Z et al. Reversing insufficient photothermal therapy-induced tumor relapse and metastasis by regulating cancer-associated fibroblasts. Nat. Comm. 13(1), 1–19 (2022). •• A intelligent study in order to boost photothermal therapy-induced antitumor immunity enough to control the growth of solid tumor and overcome the tumor immunosuppressive microenvironment.
    • 13. Kroemer G, Senovilla L, Galluzzi L, André F, Zitvogel L. Natural and therapy-induced immunosurveillance in breast cancer. Nat. Med. 21(10), 1128–1138 (2015).
    • 14. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 12(12), 860–875 (2012).
    • 15. Srivastava P. Interaction of heat shock proteins with peptides and antigen presenting cells: chaperoning of the innate and adaptive immune responses. Ann. Rev. Immunol. 20(1), 395–425 (2002).
    • 16. Multhoff G, Pockley AG, Streffer C, Gaipl US. Dual role of heat shock proteins (HSPs) in anti-tumor immunity. Curr. Mol. Med. 12(9), 1174–1182 (2012).
    • 17. Tang D, Kang R, Cheh C-W et al. HMGB1 release and redox regulates autophagy and apoptosis in cancer cells. Oncogene 29(38), 5299–5310 (2010).
    • 18. Goetz M, Toft D, Ames M, Erlichman C. The HSP90 chaperone complex as a novel target for cancer therapy. Ann. Oncol. 14(8), 1169–1176 (2003).
    • 19. Paudel YN, Angelopoulou E, Piperi C, Balasubramaniam VR, Othman I, Shaikh MF. Enlightening the role of high mobility group box 1 (HMGB1) in inflammation: updates on receptor signalling. Eur. J. Pharmacol. 858, DOI: 10.1016/j.ejphar.2019.172487 (2019).
    • 20. Gong T, Liu L, Jiang W, Zhou R. DAMP-sensing receptors in sterile inflammation and inflammatory diseases. Nat. Rev. Immunol. 20(2), 95–112 (2020).
    • 21. Kielbik M, Szulc-Kielbik I, Klink M. Calreticulin – multifunctional chaperone in immunogenic cell death: potential significance as a prognostic biomarker in ovarian cancer patients. Cells 10(1), 130 (2021).
    • 22. Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Ann. Rev. Immunol. 31, 51–72 (2013).
    • 23. Kroemer G, Galassi C, Zitvogel L, Galluzzi L. Immunogenic cell stress and death. Nat. Immunol. 23(4), 487–500 (2022). • The goal of this study was to comprehend how the immune response is affected by signals released by dying mammalian cells. The importance of immunogenic cell death in adaptive immune responses to infected or cancerous cells is emphasized, and this has significant ramifications for noninfectious, nonmalignant illnesses linked to autoreactivity.
    • 24. Podolska MJ, Shan X, Janko C et al. Graphene-induced hyperthermia (GIHT) combined with radiotherapy fosters immunogenic cell death. Front. Oncol. 11, 1–12 (2021).
    • 25. Vulpis E, Cecere F, Molfetta R et al. Genotoxic stress modulates the release of exosomes from multiple myeloma cells capable of activating NK cell cytokine production: role of HSP70/TLR2/NF-kB axis. Oncoimmunology 6(3), e1279372 (2017).
    • 26. Jella K, Nasti T, Li Z et al. Post-irradiated tumor-derived exosomes lead to melanoma tumor growth delay, potentially mediated by death associated molecular pattern (damps) proteins. Int. J. Radiat. Oncol. Biol. Phys. 102(3), S155 (2018).
    • 27. Robbins PD, Morelli AE. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 14(3), 195–208 (2014). • Extracellular vesicles are the subject of this study, which focuses on their function in intercellular communication, their contribution to immunological regulation and their potential therapeutic benefits, notably in the treatment of inflammatory illnesses, autoimmune conditions and cancer.
    • 28. Theodoraki M-N, Laban S, Jackson E et al. Changes in circulating exosome molecular profiles following surgery/(chemo) radiotherapy: early detection of response in head and neck cancer patients. Br. J. Cancer 125(12), 1677–1686 (2021). •• As plasma-derived exosomes carry immunomodulatory molecules that correlate with clinical parameters, this study explores the use of these exosomes in head and neck cancer patients as a potential biomarker for early detection of treatment failure and disease recurrence.
    • 29. Najaflou M, Shahgolzari M, Khosroushahi AY, Fiering S. Tumor-derived extracellular vesicles in cancer immunoediting and their potential as oncoimmunotherapeutics. Cancers 15(1), 82 (2022).
    • 30. Sweeney EE, Cano-Mejia J, Fernandes R. Photothermal therapy generates a thermal window of immunogenic cell death in neuroblastoma. Small 14(20), e1800678 (2018).
    • 31. Wang X, Li J, Kawazoe N, Chen G. Photothermal ablation of cancer cells by albumin-modified gold nanorods and activation of dendritic cells. Materials 12(1), 31 (2018).
    • 32. Wang C, Xu L, Liang C, Xiang J, Peng R, Liu Z. Immunological responses triggered by photothermal therapy with carbon nanotubes in combination with anti-CTLA-4 therapy to inhibit cancer metastasis. Adv. Mater. 26(48), 8154–8162 (2014).
    • 33. Huang L, Li Y, Du Y et al. Mild photothermal therapy potentiates anti-PD-L1 treatment for immunologically cold tumors via an all-in-one and all-in-control strategy. Nat. Comm. 10(1), 1–15 (2019). •• This study suggests a method known as ‘local symbiotic mild photothermal-assisted immunotherapy’ that uses photothermal therapy to make ‘cold’ tumors susceptible to immune checkpoint inhibition. This method involves loading a photothermal agent and programmed death-ligand 1 antibody into a lipid-gel depot, which increases the amount of infiltrated lymphocytes into the tumors and improves T-cell activity against tumors.
    • 34. Huang Z, Wang Y, Yao D, Wu J, Hu Y, Yuan A. Nanoscale coordination polymers induce immunogenic cell death by amplifying radiation therapy mediated oxidative stress. Nat. Comm. 12(1), 1–18 (2021).
    • 35. Li X, Lovell JF, Yoon J, Chen X. Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol. 17(11), 657–674 (2020).
    • 36. Nam J, Son S, Ochyl LJ, Kuai R, Schwendeman A, Moon JJ. Chemo-photothermal therapy combination elicits anti-tumor immunity against advanced metastatic cancer. Nat. Comm. 9(1), 1–13 (2018).
    • 37. Li W, Yang J, Luo L et al. Targeting photodynamic and photothermal therapy to the endoplasmic reticulum enhances immunogenic cancer cell death. Nat. Comm. 10(1), 1–16 (2019).
    • 38. Ali MR, Snyder B, El-Sayed MA. Synthesis and optical properties of small Au nanorods using a seedless growth technique. Langmuir 28(25), 9807–9815 (2012).
    • 39. Tebbe M, Kuttner C, Männel M, Fery A, Chanana M. Colloidally stable and surfactant-free protein-coated gold nanorods in biological media. ACS Appl. Mater. Interfaces 7(10), 5984–5991 (2015).
    • 40. Richardson HH, Carlson MT, Tandler PJ, Hernandez P, Govorov AO. Experimental and theoretical studies of light-to-heat conversion and collective heating effects in metal nanoparticle solutions. Nano Letters 9(3), 1139–1146 (2009).
    • 41. Ge J, Jia Q, Liu W et al. Red-emissive carbon dots for fluorescent, photoacoustic, and thermal theranostics in living mice. Adv. Mater. 27(28), 4169–4177 (2015).
    • 42. Théry C, Amigorena S, Raposo G, Clayton A. Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr. Protocol. Cell Biol. 30(1), 3.22.21–3.22.29 (2006).
    • 43. Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc. Natl Acad. Sci. USA 76(9), 4350–4354 (1979).
    • 44. Shahgolzari M, Pazhouhandeh M, Milani M, Fiering S, Khosroushahi AY. Alfalfa mosaic virus nanoparticles-based in situ vaccination induces antitumor immune responses in breast cancer model. Nanomedicine 16(2), 97–107 (2021).
    • 45. Berne BJ, Pecora R. Dynamic Light Scattering: With Applications to Chemistry, Biology, and Physics. Courier Corporation, Washington, DC, USA (2000).
    • 46. Arenas-Guerrero P, Delgado ÁV, Donovan KJ et al. Determination of the size distribution of non-spherical nanoparticles by electric birefringence-based methods. Sci. Rep. 8(1), 9502 (2018).
    • 47. Reimer L. Transmission Electron Microscopy: Physics of Image Formation and Microanalysis. Springer, New York, NY, USA, 36 (2013).
    • 48. Clogston JD, Patri AK. Zeta potential measurement. Methods Mol. Biol. 697, 63–70 (2011).
    • 49. James ML, Gambhir SS. A molecular imaging primer: modalities, imaging agents, and applications. Physiol. Rev. 92(2), 897–965 (2012).
    • 50. Zhang JZ, Noguez C. Plasmonic optical properties and applications of metal nanostructures. Plasmonics 3(4), 127–150 (2008).
    • 51. Qin Z, Wang Y, Randrianalisoa J et al. Quantitative comparison of photothermal heat generation between gold nanospheres and nanorods. Sci. Rep. 6(1), 29836 (2016).
    • 52. Gamrekelashvili J, Ormandy LA, Heimesaat MM et al. Primary sterile necrotic cells fail to cross-prime CD8+ T cells. Oncoimmunology 1(7), 1017–1026 (2012).
    • 53. Li Y, He L, Dong H et al. Fever-inspired immunotherapy based on photothermal CpG nanotherapeutics: the critical role of mild heat in regulating tumor microenvironment. Adv. Sci. 5(6), doi: 10.1002/advs.201700805 (2018). •• A photothermal CpG nanotherapeutics approach is introduced that employs low-intensity heat to create an immunofavorable tumor microenvironment, resulting in improved antitumor immune effects, activated immune responses and apoptotic cell death.
    • 54. Taylor ML, Wilson RE Jr, Amrhein KD, Huang X. Gold nanorod-assisted photothermal therapy and improvement strategies. Bioengineering 9(5), 200 (2022).
    • 55. Ali MR, Rahman MA, Wu Y et al. Efficacy, long-term toxicity, and mechanistic studies of gold nanorods photothermal therapy of cancer in xenograft mice. Proc. Natl Acad. Sci. USA 114(15), E3110–E3118 (2017).
    • 56. Kannadorai RK, Liu Q. Optimization in interstitial plasmonic photothermal therapy for treatment planning. Med. Phys. 40(10), 103301 (2013).
    • 57. Dickerson EB, Dreaden EC, Huang X et al. Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Lett. 269(1), 57–66 (2008).
    • 58. Kang X, Guo X, Niu X et al. Photothermal therapeutic application of gold nanorods-porphyrin-trastuzumab complexes in HER2-positive breast cancer. Sci. Rep. 7(1), 42069 (2017).
    • 59. Schachter D. The Source of Toxicity in CTAB and CTAB-Stabilized Gold Nanorods. Rutgers, The State University of New Jersey, NJ, USA (2013).
    • 60. Leroueil PR, Hong S, Mecke A, Baker JR Jr, Orr BG, Banaszak Holl MM. Nanoparticle interaction with biological membranes: does nanotechnology present a Janus face? Acc. Chem. Res. 40(5), 335–342 (2007).
    • 61. Majno G, Joris I. Apoptosis, oncosis, and necrosis. An overview of cell death. Am. J. Pathol. 146(1), 3 (1995).
    • 62. Wang L, Liu Y, Li W et al. Selective targeting of gold nanorods at the mitochondria of cancer cells: implications for cancer therapy. Nano Lett. 11(2), 772–780 (2011).
    • 63. Morlé A, Garrido C, Micheau O. Hyperthermia restores apoptosis induced by death receptors through aggregation-induced c-FLIP cytosolic depletion. Cell Death Dis. 6(2), e1633–e1633 (2015).
    • 64. Hildebrandt B, Wust P, Ahlers O et al. The cellular and molecular basis of hyperthermia. Crit. Rev. Oncol./Hematol. 43(1), 33–56 (2002).
    • 65. De Toro J, Herschlik L, Waldner C, Mongini C. Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications. Front. Immunol. 6, 203 (2015).
    • 66. Cho J-A, Lee Y-S, Kim S-H, Ko J-K, Kim C-W. MHC independent anti-tumor immune responses induced by HSP70-enriched exosomes generate tumor regression in murine models. Cancer Lett. 275(2), 256–265 (2009).
    • 67. Komarova EY, Suezov RV, Nikotina AD et al. HSP70-containing extracellular vesicles are capable of activating of adaptive immunity in models of mouse melanoma and colon carcinoma. Sci. Rep. 11(1), 1–16 (2021).
    • 68. Khandelwal A, Kent CN, Balch M et al. Structure-guided design of an HSP90β N-terminal isoform-selective inhibitor. Nat. Comm. 9(1), 1–7 (2018).
    • 69. Wang H, Bloom O, Zhang M et al. HMG-1 as a late mediator of endotoxin lethality in mice. Science 285(5425), 248–251 (1999).
    • 70. Tugues S, Burkhard S, Ohs IA et al. New insights into IL-12-mediated tumor suppression. Cell Death Different 22(2), 237–246 (2015).
    • 71. Tulotta C, Lefley DV, Moore CK et al. IL-1B drives opposing responses in primary tumours and bone metastases; harnessing combination therapies to improve outcome in breast cancer. NPJ Breast Cancer 7(1), 1–15 (2021).
    • 72. Chen T, Guo J, Yang M, Zhu X, Cao X. Chemokine-containing exosomes are released from heat-stressed tumor cells via lipid raft-dependent pathway and act as efficient tumor vaccine. J. Immunol. 186(4), 2219–2228 (2011).
    • 73. Guo D, Chen Y, Wang S et al. Exosomes from heat-stressed tumour cells inhibit tumour growth by converting regulatory T cells to Th17 cells via IL-6. Immunology 154(1), 132–143 (2018).
    • 74. Loi S, Drubay D, Adams S et al. Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers. J. Clin. Oncol. 37(7), 559 (2019).
    • 75. Liu Y-T, Sun Z-J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 11(11), 5365 (2021). • Discusses various strategies, including nanomedicines, to convert cold tumors into hot tumors with increased T-cell infiltration and improved immune checkpoint inhibitor efficacy, suggesting the potential for multiple T cell-based combination therapies.