Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1887 2022-05-20 08:34:48 |
2 layout Meta information modification 1887 2022-05-20 08:49:37 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Li, J.; , .; Wang, Y. Nanomedicines for NIR-II Photothermal Therapy Combinational Immunotherapy. Encyclopedia. Available online: https://encyclopedia.pub/entry/23154 (accessed on 27 July 2024).
Li J,  , Wang Y. Nanomedicines for NIR-II Photothermal Therapy Combinational Immunotherapy. Encyclopedia. Available at: https://encyclopedia.pub/entry/23154. Accessed July 27, 2024.
Li, Jingchao, , Yongtao Wang. "Nanomedicines for NIR-II Photothermal Therapy Combinational Immunotherapy" Encyclopedia, https://encyclopedia.pub/entry/23154 (accessed July 27, 2024).
Li, J., , ., & Wang, Y. (2022, May 20). Nanomedicines for NIR-II Photothermal Therapy Combinational Immunotherapy. In Encyclopedia. https://encyclopedia.pub/entry/23154
Li, Jingchao, et al. "Nanomedicines for NIR-II Photothermal Therapy Combinational Immunotherapy." Encyclopedia. Web. 20 May, 2022.
Nanomedicines for NIR-II Photothermal Therapy Combinational Immunotherapy
Edit

Photothermal therapy (PTT) utilizes the light irradiation of photothermal agents to generate heat for cancer cell killing. PTT has shown a great promise for cancer treatment because of the noninvasiveness, high spatiotemporal precision, simple operation, and flexible tenability of light sources.

second near-infrared windows immunotherapy optical nanomedicines

1. Introduction

In recent years, immunotherapy has become another effective treatment strategy for cancer after surgery, radiotherapy, chemotherapy, and targeted therapy [1][2][3]. Immunotherapy that trains the host antitumor immune responses can eliminate both local tumors and distant metastases, as well as trigger long-term immune memory to prevent tumor recurrence [4][5][6]. Currently, cancer vaccines, chimeric antigen receptor T-cell therapy, and immune checkpoint blockade therapy are the three main strategies for immunotherapy [7][8][9]. A large number of immunotherapeutic drugs have been approved for the treatment of different malignant tumors, leading to effective tumor treatment in a subset of patients [10][11][12][13]. However, there are two major challenges for clinical applications of immunotherapy [14][15][16]. First, the response rates of patients for immunotherapy are low, which results in limited therapeutic efficacy [17][18][19]. For example, only around 10–30% tumors in clinical patients can be effectively treated by immune checkpoint blockers, while most of the tumors respond poorly to immune checkpoint blockade therapy because of their low immunogenicity [20][21][22][23]. Second, immunotherapy has the possibility to cause immune-related adverse events, such as diabetes mellitus, myocarditis, thyroid dysfunction, hypophysitis, and hypokalemia, particularly for high-dosage injections and/or combinations of multiple immunotherapeutic drugs [24][25][26][27][28]. Therefore, it is highly desired to develop effective and safe approaches to achieve ideal antitumor immune responses.
Photothermal therapy (PTT) utilizes the light irradiation of photothermal agents to generate heat for cancer cell killing. PTT has shown a great promise for cancer treatment because of the noninvasiveness, high spatiotemporal precision, simple operation, and flexible tenability of light sources [29][30][31][32]. Thus, PTT often shows high selectivity and specificity for cancer treatment without causing obvious damage to normal tissues [33][34][35][36]. The main light sources used for PTT are the first NIR light (NIR-I, 650–950 nm), which however, has limited tissue penetration depth (less than 1 cm) and relatively low maximum permissible exposure for skin [37][38][39]. Therefore, the extensive applications of PTT have been greatly hindered. To address these issues, a new optical window termed the second NIR (NIR-II) window (1000–1700 nm), with better tissue penetration depth (around 3–5 cm) and higher maximum permissible exposure relative to NIR-II light, has been adopted for NIR-II PTT [40][41][42]. Such advantages of NIR-II light not only enable the treatment of deep regions of tumors but also allow for strong power density to improve heat generation, thus achieving high PTT therapeutic efficacy [43]. To date, different nanosystems such as metallic nanoparticles, metal–organic hybrid nanoparticles, inorganic semiconducting nanoparticles, organic polymer nanoparticles, and small-molecule-based nanoparticles have been developed for NIR-II PTT [44][45][46][47][48].
In addition to the direct ablation of tumors, NIR-II PTT has been recently used to reprogram the tumor immunosuppressive microenvironment to potentiate cancer immunotherapy [49]. The generated heat during NIR-II PTT can induce immunogenic cell death (ICD) of cancer cells, which is characterized by the release of tumor-associated antigens, adenosine triphosphate, and high mobility group box 1 protein into the extracellular environment, and the translocation of calreticulin to the cell surface [19]. Such an action can promote the uptake and processing of antigens by antigen presentation cells and facilitate the production and priming of effector T cells, leading to the activation of antitumor immunity for the eradication of tumors and metastases [50]. In addition, the NIR-II PTT-mediated ICD effect can further enhance antitumor immune responses of immunotherapy, leading to combinational action for effective treatments of tumors [51].

2. Organic Nanomedicines for NIR-II PTT Combinational Immunotherapy

2.1. Small-Molecule-Based Nanoparticles

Some organic small-molecule-based nanoparticles with strong NIR-II absorbance and excellent photothermal conversion efficacy have been constructed for NIR-II PTT combinational immunotherapy. In a recent study of researchers' group, 3,3′,5,5′-tetramethylbenzidine (TMB)-based liposome nanocomplexes with pH-responsive NIR-II photothermal properties were constructed for combinational immunotherapy [52]. Thermal-responsive liposomes containing an amphiphilic polymer, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol) (DSPE-PEG), and thermal-responsive 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), with a transition temperature of 41 °C, were synthesized to load pH-sensitive charge-transfer nanoparticles as the NIR-II photothermal agent, deoxyribonuclease I (DNase I), and stimulants of the natural killer (NK) cell (SIS3). Charge-transfer nanoparticles were transformed from TMB and exhibited a stronger absorption in the NIR-II window in an acidic environment relative to neutral and basic conditions and thus have pH-sensitive NIR-II photothermal properties [53]. Such nanocomplexes exerted a NIR-II PTT effect under 1064 nm laser irradiation (1.0 W/cm2, 6 min) in a tumor acidic microenvironment, resulting in the increase of tumor temperature at around 45 °C. This destroyed the thermal-responsive liposomes to allow the on-demand release of SIS3 and DNase I. Due to the toxicity of DNase I, cancer cells were killed, and ICD was induced to promote immune responses. The action of SIS3 could synergize with DNase I-mediated ICD to promote the activation of NK cells and CD8+ cytotoxic T lymphocytes (CTLs). Thus, such a combinational immunotherapy could effectively inhibit the growth of primary and distant 4T1 tumors and completely prevented lung metastasis in subcutaneous mouse models.
The combination of reactive oxygen species (ROS) and NIR-II PTT-mediated heat generation for ICD induction and the promotion of immunotherapy has also been reported. As shown in Zhao’s group, small-molecule-based organic metal adjuvants (OMAs) with NIR-II photothermal properties and ROS-generating ability were developed for cancer therapy [54]. Such OMAs were constructed through the supramolecular assembly of commercially available donors and acceptors, showing excellent NIR-II photothermal properties and photoacoustic imaging performance via optimizing the constituting components. In a tumor microenvironment, OMAs oxidized cysteine and glutathione (GSH) to inhibit the biosynthesis of GSH, resulting in the disruption of redox homeostasis and boosting ROS accumulation inside cells. Under 1064 nm laser irradiation (1.0 W/cm2, 5 min), OMAs exerted an NIR-II PTT effect to ablate cancer cells. In addition, the PTT effect and ROS generation mediated ICD induction with improved efficacy to enhance the immune response by promoting the maturation of dendritic cells (DCs) and the infiltration of T cells. Such an ROS-generating NIR-II PTT could be combined with anti-programmed cell death protein 1 (aPD-1) antibody-mediated immune checkpoint blockade therapy to allow for the increased infiltration of T cells into tumor tissues, leading to the eradication of primary tumors and the significant inhibition of distant tumor growth in 4T1 tumor-bearing-mouse models.
To boost ROS generation for combining NIR-II PTT and immunotherapy, Shen’s group reported an “all-in-one” hydrogel for cancer treatment [55]. Such hydrogels were formed by loading ink as the NIR-II photothermal agent, HY19991 as the PD-L1 inhibitor, and an azo-initiator of 2,2-azobis [2-(2-imidazoline-2-acyl)propane]dihydrochloride (AIPH) into alginate hydrogels in situ crosslinked with Ca2+. Under 1064 nm laser irradiation (0.5 W/cm2, 10 min), the ink exerted NIR-II PTT to increase the local temperature at around 45 °C, leading to the upregulation of PD-L1 expression and the formation of a large number of alkyl radicals from AIPH. The formed alkyl radicals augmented the ICD effect and increased the recruitment of tumor-infiltrating lymphocytes into tumors via the hydrogel-mediated conversion of “cold” tumors into “hot” tumors. Moreover, hydrogels released HY19991 to block the binding between PD-L1 and PD-1 to further improve the antitumor immunity. As a result, such “all-in-one” hydrogels could afford synergistic action via a mild PTT effect to reverse the tumor immunosuppressive microenvironment and triggered both innate and adaptive immune responses in CT26 tumor-bearing-mouse models, leading to the effective elimination of tumors and the prevention of distant metastatic tumors.

2.2. Semiconducting Polymer Nanoparticles (SPNs)

SPNs, as a class of optical materials transformed from semiconducting polymers (SPs) with excellent optical properties and biocompatibility, have also been used for NIR-II PTT and thus can mediate NIR-II PTT combinational immunotherapy [56][57][58]. To enhance the therapeutic efficacy of SPNs, Zhang and Pu’s groups developed a polymer multicellular nanoengager for synergistic NIR-II photothermal immunotherapy [59]. The nanoengager consisted of NIR-II-absorbing SPs as the photothermal agents and the surface camouflaged cell membranes derived from DCs and immunologically engineered tumor cells serving as the cancer vaccine shells. Such a design enabled multicellular engagements among T cells, DCs, and tumor cells, resulting in the enhanced activation of DCs and T cells. These nanoengagers could effectively accumulate into both lymph nodes and tumor tissues after systemic administration and acted as nanovaccines to trigger the immune response. Under 1064 nm laser irradiation (1.0 W/cm2, 10 min), the nanoengagers exerted NIR-II PTT to eradicate tumors and induce ICD for further eliciting antitumor T cell immunity. The nanoengager-mediated NIR-II PTT synergistic immunotherapy not only efficiently inhibited the growth of both primary and distant 4T1 tumors and eliminated tumor recurrence but also triggered immunological memory for long-term immune surveillance.
By using thermo-responsive linkers, Pu’s groups reported an activatable polymer nanoagonist for the NIR-II photothermal immunotherapy of cancer [60]. The nanoagonists were constructed by covalently conjugating R848 onto NIR-II-absorbing SPNs via a labile thermo-responsive linker. Under 1064 nm laser irradiation (1.0 W/cm2, 10 min), the nanoagonists exerted NIR-II PTT for killing tumor cells and inducing ICD. The generated heat also destroyed thermo-responsive linkers to achieve the on-demand release of R848 even in deep solid tumor tissue. The antitumor immune response in 4T1 tumor-bearing-mouse models was potentiated due to the combinational action of the NIR-II PTT-mediated ICD effect and R848-mediated TLR activation. Therefore, through the combinational action of NIR-II PTT and immunotherapy, these nanoagonists not only almost completely eradicated the primary tumors after direct laser irradiation but also obviously inhibited the growth of distant tumors and suppressed lung and liver metastasis.
In another study, Pu and coworkers reported a NIR-II light-activatable polymeric nanoantagonist for photothermal immunometabolic cancer therapy [61]. The polymeric nanoantagonist was obtained by conjugating an adenosine A2A receptor antagonist (vipadenant) onto NIR-II-absorbing SPs via the thermo-responsive linkers. Under 1064 nm laser irradiation (1.0 W/cm2, 10 min), SPNs within nanoantagonists mediated NIR-II PTT to induce tumor thermal ablation and subsequently ICD, and the release of vipadenant through triggering the cleavage of the thermo-responsive linkers. The released vipadenant could block the binding between extracellular adenosine with A2A receptors on the surface of the CTLs and the regulatory T (Treg) cells, thus promoting the priming and infiltration of CTLs but suppressing the functions of the Treg cells to achieve an enhanced antitumor immune response. Such a combinational action of NIR-II PTT and immunotherapy, which was mediated by these nanoantagonists, allowed for the complete eradication of primary 4T1 tumors, the effective inhibition of metastasis, and the prevention of tumor relapse after reinoculation.

3. Inorganic Non-Metal Nanomedicines for NIR-II PTT Combinational Immunotherapy

Some inorganic non-metal nanomaterials possessing an intrinsic nature of strong absorbance in the NIR-II window and good photothermal conversion efficacy can produce heat under NIR-II laser irradiation and thus are extensively expanded to manage the synergistic tumor treatment of PTT and immunotherapy. Xing’s group developed an immunoadjuvant-modified nanotube platform to accomplish the NIR-II PTT combinational immunotherapy of mouse tumors [62]. Single-walled carbon nanotubes (SWNTs) prepared by exfoliating pristine SWNTs under an ultrasonic process were mixed with glycated chitosan (GC) to construct the immunological SWNT-GC nanoplatforms. SWNT-GC not only maintained the optical and photothermal properties of SWNTs and the immunological functions of GC but also could be easily internalized into cells for efficient PTT treatment and triggering an immune response under NIR-II laser irradiation (0.75 W/cm2, 10 min). For tumor immunogenicity, GC acted as damage-associated molecular pattern molecules (DAMPs) and pathogen-associated molecular pattern molecules (PAMPs) for the enhanced presentation of antigens to reinforce the antitumor immunity. SWNT-GC treatment with laser irradiation afforded remarkably improved efficacy in suppressing tumor growth, increasing long-term mouse survival, and inhibiting tumor rechallenge using EMT6 tumor-bearing-mouse models.

References

  1. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264.
  2. Vanneman, M.; Dranoff, G. Combining immunotherapy and targeted therapies in cancer treatment. Nat. Rev. Cancer 2012, 12, 237–251.
  3. Riley, R.S.; June, C.H.; Langer, R.; Mitchell, M.J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175–196.
  4. Mellman, I.; Coukos, G.; Dranoff, G. Cancer immunotherapy comes of age. Nature 2011, 480, 480–489.
  5. Wang, C.; Sun, W.; Ye, Y.; Hu, Q.; Bomba, H.N.; Gu, Z. In situ activation of platelets with checkpoint inhibitors for post-surgical cancer immunotherapy. Nat. Biomed. Eng. 2017, 1, 1–10.
  6. Tang, L.; Zheng, Y.; Melo, M.B.; Mabardi, L.; Castaño, A.P.; Xie, Y.-Q.; Li, N.; Kudchodkar, S.B.; Wong, H.C.; Jeng, E.K.; et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 2018, 36, 707–716.
  7. Chen, F.; Wang, Y.; Gao, J.; Saeed, M.; Li, T.; Wang, W.; Yu, H. Nanobiomaterial-based vaccination immunotherapy of cancer. Biomaterials 2021, 270, 120709.
  8. Neelapu, S.S.; Tummala, S.; Kebriaei, P.; Wierda, W.; Gutierrez, C.; Locke, F.L.; Komanduri, K.V.; Lin, Y.; Jain, N.; Daver, N. Chimeric antigen receptor T-cell therapy—Assessment and management of toxicities. Nat. Rev. Clin. Oncol. 2018, 15, 47–62.
  9. Byun, D.J.; Wolchok, J.D.; Rosenberg, L.M.; Girotra, M. Cancer immunotherapy—Immune checkpoint blockade and associated endocrinopathies. Nat. Rev. Endocrinol. 2017, 13, 195–207.
  10. Fan, Q.; Chen, Z.; Wang, C.; Liu, Z. Toward biomaterials for enhancing immune checkpoint blockade therapy. Adv. Funct. Mater. 2018, 28, 1802540.
  11. Ng, C.W.; Li, J.; Pu, K. Recent progresses in phototherapy-synergized cancer immunotherapy. Adv. Funct. Mater. 2018, 28, 1804688.
  12. Feng, B.; Zhou, F.; Hou, B.; Wang, D.; Wang, T.; Fu, Y.; Ma, Y.; Yu, H.; Li, Y. Binary cooperative prodrug nanoparticles improve immunotherapy by synergistically modulating immune tumor microenvironment. Adv. Mater. 2018, 30, 1803001.
  13. Konstantinidou, M.; Zarganes-Tzitzikas, T.; Magiera-Mularz, K.; Holak, T.A.; Dömling, A. Immune checkpoint PD-1/PD-L1. Is there life beyond antibodies? Angew. Chem. Int. Ed. 2018, 57, 4840–4848.
  14. Li, J.; Cui, D.; Huang, J.; He, S.; Yang, Z.; Zhang, Y.; Luo, Y.; Pu, K. Organic semiconducting pro-nanostimulants for near-infrared photoactivatable cancer immunotherapy. Angew. Chem. Int. Ed. 2019, 58, 12680–12687.
  15. Li, J.; Luo, Y.; Pu, K. Electromagnetic nanomedicines for combinational cancer immunotherapy. Angew. Chem. Int. Ed. 2021, 60, 12682–12705.
  16. Zeng, Z.; Zhang, C.; Li, J.; Cui, D.; Jiang, Y.; Pu, K. Activatable polymer nanoenzymes for photodynamic immunometabolic cancer therapy. Adv. Mater. 2021, 33, 2007247.
  17. Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218.
  18. Goldberg, M.S. Improving cancer immunotherapy through nanotechnology. Nat. Rev. Cancer 2019, 19, 587–602.
  19. Shi, Y.; Lammers, T. Combining nanomedicine and immunotherapy. Acc. Chem. Res. 2019, 52, 1543–1554.
  20. Topalian, S.L.; Taube, J.M.; Anders, R.A.; Pardoll, D.M. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Rev. Cancer 2016, 16, 275–287.
  21. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571.
  22. Nam, J.; Son, S.; Park, K.S.; Zou, W.; Shea, L.D.; Moon, J.J. Cancer nanomedicine for combination cancer immunotherapy. Nat. Rev. Mater. 2019, 4, 398–414.
  23. Irvine, D.J.; Dane, E.L. Enhancing cancer immunotherapy with nanomedicine. Nat. Rev. Immunol. 2020, 20, 321–334.
  24. Boutros, C.; Tarhini, A.; Routier, E.; Lambotte, O.; Ladurie, F.L.; Carbonnel, F.; Izzeddine, H.; Marabelle, A.; Champiat, S.; Berdelou, A. Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat. Rev. Clin. Oncol. 2016, 13, 473–486.
  25. Sullivan, R.J.; Weber, J.S. Immune-related toxicities of checkpoint inhibitors. mechanisms and mitigation strategies. Nat. Rev. Drug Discov. 2021, 20, 427–453.
  26. Shen, S.; Dai, H.; Fei, Z.; Chai, Y.; Hao, Y.; Fan, Q.; Dong, Z.; Zhu, Y.; Xu, J.; Ma, Q. Immunosuppressive nanoparticles for management of immune-related adverse events in liver. ACS Nano 2021, 15, 9111–9125.
  27. Hansel, T.T.; Kropshofer, H.; Singer, T.; Mitchell, J.A.; George, A.J. The safety and side effects of monoclonal antibodies. Nat. Rev. Drug Discov. 2010, 9, 325–338.
  28. Weiden, J.; Tel, J.; Figdor, C.G. Synthetic immune niches for cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 212–219.
  29. Jung, H.S.; Verwilst, P.; Sharma, A.; Shin, J.; Sessler, J.L.; Kim, J.S. Organic molecule-based photothermal agents. an expanding photothermal therapy universe. Chem Soc. Rev. 2018, 47, 2280–2297.
  30. Liu, Y.; Bhattarai, P.; Dai, Z.; Chen, X. Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer. Chem. Soc. Rev. 2019, 48, 2053–2108.
  31. Hu, J.-J.; Cheng, Y.-J.; Zhang, X.-Z. Recent advances in nanomaterials for enhanced photothermal therapy of tumors. Nanoscale 2018, 10, 22657–22672.
  32. Zhao, J.; Cui, W. Functional electrospun fibers for local therapy of cancer. Adv. Fiber. Mater. 2020, 2, 229–245.
  33. Zheng, B.-D.; He, Q.-X.; Li, X.; Yoon, J.; Huang, J.-D. Phthalocyanines as contrast agents for photothermal therapy. Coord. Chem. Rev. 2021, 426, 213548.
  34. Li, J.; Pu, K. Semiconducting polymer nanomaterials as near-infrared photoactivatable protherapeutics for cancer. Acc. Chem. Res. 2020, 53, 752–762.
  35. Li, J.; Pu, K. Development of organic semiconducting materials for deep-tissue optical imaging, phototherapy and photoactivation. Chem. Soc. Rev. 2019, 48, 38–71.
  36. Li, J.; Yu, X.; Shi, X.; Shen, M. Cancer nanomedicine based on polyethylenimine-mediated multifunctional nanosystems. Prog. Mater. Sci. 2022, 124, 100871.
  37. Jiang, Y.; Zhao, X.; Huang, J.; Li, J.; Upputuri, P.K.; Sun, H.; Han, X.; Pramanik, M.; Miao, Y.; Duan, H. Transformable hybrid semiconducting polymer nanozyme for second near-infrared photothermal ferrotherapy. Nat. Commun. 2020, 11, 1857.
  38. Hong, G.; Antaris, A.L.; Dai, H. Near-infrared fluorophores for biomedical imaging. Nat. Biomed. Eng. 2017, 1, 0010.
  39. Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nat. Mater. 2013, 12, 991–1003.
  40. Jiang, Y.; Li, J.; Zhen, X.; Xie, C.; Pu, K. Dual-peak absorbing semiconducting copolymer nanoparticles for first and second near-infrared window photothermal therapy. a comparative study. Adv. Mater. 2018, 30, 1705980.
  41. Lin, H.; Gao, S.; Dai, C.; Chen, Y.; Shi, J. A two-dimensional biodegradable niobium carbide (MXene) for photothermal tumor eradication in NIR-I and NIR-II biowindows. J. Am. Chem. Soc. 2017, 139, 16235–16247.
  42. Guo, B.; Sheng, Z.; Hu, D.; Liu, C.; Zheng, H.; Liu, B. Through scalp and skull NIR-II photothermal therapy of deep orthotopic brain tumors with precise photoacoustic imaging guidance. Adv. Mater. 2018, 30, 1802591.
  43. Lyu, Y.; Li, J.; Pu, K. Second near-infrared absorbing agents for photoacoustic imaging and photothermal therapy. Small Methods 2019, 3, 1900553.
  44. Chen, Y.-S.; Zhao, Y.; Yoon, S.J.; Gambhir, S.S.; Emelianov, S. Miniature gold nanorods for photoacoustic molecular imaging in the second near-infrared optical window. Nat. Nanotechnol. 2019, 14, 465–472.
  45. Zhang, D.; Xu, H.; Zhang, X.; Liu, Y.; Wu, M.; Li, J.; Yang, H.; Liu, G.; Liu, X.; Liu, J. Self-quenched metal-organic particles as dual-mode therapeutic agents for photoacoustic imaging-guided second near-infrared window photochemotherapy. ACS Appl. Mater. Interfaces 2018, 10, 25203–25212.
  46. Guo, C.; Yu, H.; Feng, B.; Gao, W.; Yan, M.; Zhang, Z.; Li, Y.; Liu, S. Highly efficient ablation of metastatic breast cancer using ammonium-tungsten-bronze nanocube as a novel 1064 nm-laser-driven photothermal agent. Biomaterials 2015, 52, 407–416.
  47. Cao, Z.; Feng, L.; Zhang, G.; Wang, J.; Shen, S.; Li, D.; Yang, X. Semiconducting polymer-based nanoparticles with strong absorbance in NIR-II window for in vivo photothermal therapy and photoacoustic imaging. Biomaterials 2018, 155, 103–111.
  48. Zhang, L.; Forgham, H.; Huang, X.; Shen, A.; Davis, T.; Qiao, R.; Guo, B. All-in-one inorganic nanoagents for near-infrared-II photothermal-based cancer theranostics. Mater. Today Adv. 2022, 14, 100226.
  49. Sun, H.; Zhang, Q.; Li, J.; Peng, S.; Wang, X.; Cai, R. Near-infrared photoactivated nanomedicines for photothermal synergistic cancer therapy. Nano Today 2021, 37, 101073.
  50. Duan, X.; Chan, C.; Lin, W. Nanoparticle-mediated immunogenic cell death enables and potentiates cancer immunotherapy. Angew. Chem. Int. Ed. 2019, 58, 670–680.
  51. Xu, C.; Pu, K. Second near-infrared photothermal materials for combinational nanotheranostics. Chem. Soc. Rev. 2021, 50, 1111–1137.
  52. Chen, S.; Wang, X.; Lin, M.; Hou, Y.; Ding, M.; Kong, D.; Sun, H.; Zhang, Q.; Li, J.; Zhou, Q. Liposome-based nanocomplexes with pH-sensitive second near-infrared photothermal property for combinational immunotherapy. Appl. Mater. Today 2021, 25, 101258.
  53. Wang, Z.; Upputuri, P.K.; Zhen, X.; Zhang, R.; Jiang, Y.; Ai, X.; Zhang, Z.; Hu, M.; Meng, Z.; Lu, Y. pH-sensitive and biodegradable charge-transfer nanocomplex for second near-infrared photoacoustic tumor imaging. Nano Res. 2019, 12, 49–55.
  54. Chen, Y.; He, P.; Jana, D.; Wang, D.; Wang, M.; Yu, P.; Zhu, W.; Zhao, Y. Glutathione-depleting organic metal adjuvants for effective NIR-II photothermal immunotherapy. Adv. Mater. 2022, 34, 2201706.
  55. Ning, B.; Liu, Y.; Ouyang, B.; Curation, X.S.D.; Guo, H.; Pang, Z.; Shen, S. Low-temperature photothermal irradiation triggers alkyl radicals burst for potentiating cancer immunotherapy. J. Colloid Interface Sci. 2022, 614, 436–450.
  56. Zhang, W.; Sun, X.; Huang, T.; Pan, X.; Sun, P.; Li, J.; Zhang, H.; Lu, X.; Fan, Q.; Huang, W. 1300 nm absorption two-acceptor semiconducting polymer nanoparticles for NIR-II photoacoustic imaging system guided NIR-II photothermal therapy. Chem. Commun. 2019, 55, 9487–9490.
  57. Zhen, X.; Pu, K.; Jiang, X. Photoacoustic imaging and photothermal therapy of semiconducting polymer nanoparticles. signal amplification and second near-infrared construction. Small 2021, 17, 2004723.
  58. Sun, T.; Han, J.; Liu, S.; Wang, X.; Wang, Z.Y.; Xie, Z. Tailor-made semiconducting polymers for second near-infrared photothermal therapy of orthotopic liver cancer. ACS Nano 2019, 13, 7345–7354.
  59. Xu, C.; Jiang, Y.; Han, Y.; Pu, K.; Zhang, R. A polymer multicellular nanoengager for synergistic NIR-II photothermal immunotherapy. Adv. Mater. 2021, 33, 2008061.
  60. Jiang, Y.; Huang, J.; Xu, C.; Pu, K. Activatable polymer nanoagonist for second near-infrared photothermal immunotherapy of cancer. Nat. Commun. 2021, 12, 742.
  61. Xu, C.; Jiang, Y.; Huang, J.; Huang, J.; Pu, K. Second near-infrared light-activatable polymeric nanoantagonist for photothermal immunometabolic cancer therapy. Adv. Mater. 2021, 33, 2101410.
  62. Zhou, F.; Wu, S.; Song, S.; Chen, W.R.; Resasco, D.E.; Xing, D. Antitumor immunologically modified carbon nanotubes for photothermal therapy. Biomaterials 2012, 33, 3235–3242.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 338
Revisions: 2 times (View History)
Update Date: 20 May 2022
1000/1000
Video Production Service