Introduction

Immunotherapy, which activates immune systems to combat cancer, has yielded considerable clinical benefits recently [1]. Antitumor immunotherapy can be typically divided into two categories based on the mechanism: (i) immune-enhancing therapies that reinforce immune responses against tumors, such as cytokines, vaccines, and adoptive cell therapy, and (ii) immune-normalizing therapies that repair the defects of systemic antitumor immunity, such as immune checkpoint blockade (ICB) [2]. Despite wide application in the clinic, how to obtain long-lasting responses in the majority of patients suffering cancer remains an unsolved problem for cancer immunotherapy [3].

Severe immune-related side effects (irSEs) and a low response rate hinder the progress of immunotherapy [4, 5]. The irSEs (e.g., myocarditis and pneumonia) result from nonspecific activation of the immune system due to the extratumoral distribution of the drug after systemic administration. Primary and acquired resistance in tumors results in the ineffectiveness of immunotherapy [6]. For instance, the objective response rates of programmed death 1 (PD-1) protein or its ligand (PD-L1) inhibitors against certain cancers (e.g., pancreatic cancer and glioblastoma) are lower than 30% [7]. The immunosuppressive tumor microenvironment (ITME) is a major hurdle to immunotherapy, as it supports tumor evasion of immune surveillance [8]. Therefore, develo** strategies to reverse the ITME is necessary for improving cancer immunotherapy [9, 10].

Nanosized drug delivery systems (NDDSs) improve the safety and efficacy of cancer immunotherapy because of their excellent pharmacokinetic and biodistribution profiles, including prolonged blood half-life, high intratumoral accumulation, and deep tumor penetration capacity [11,12,13]. They can also be designed as multidrug delivery platforms for combined therapy, which is beneficial for immunotherapy since ITME is an intricate network that requires modulation from multiple aspects [14,15,16].

Recently, our group reported a series of NDDSs for remodeling the ITME and enhancing cancer immunotherapy. In this review, we briefly introduce the composition of ITME and traditional treatments against ITME. Then, we will elaborate our endeavors in reversing the ITME to improve immunotherapy by manipulating NDDSs based on different strategies. Finally, the contributions and prospects of this field will be discussed.

Immunosuppressive tumor microenvironment

Characterization and composition

The tumor microenvironment (TME) is highly heterogeneous among tumors [17]. Based on the type, density and location of immune cells within the tumor site, the tumor immune microenvironment can be broadly classified into two categories: hot and cold tumors [18]. Immunologically hot tumors, with high infiltration of cytotoxic T lymphocytes (CTLs) and activation of the PD-1/PD-L1 signaling pathway, are responsive to immunotherapy [19]. Unfortunately, many tumors insensitive to immunotherapy are usually cold tumors with ITME, which are difficult to eradicate and associated with poor prognosis [20].

Multiple and complex factors contribute to the ITME. The low mutational burden and poor immunogenicity of tumors prevent recognition by the immune system [21, 22]. Various immunosuppressive cells and cytokines impede antitumor immune responses through different signaling pathways [23]. The extracellular matrix and chemokines of tumors block the penetration of antitumor immune cells [24, 25]. Both the physicochemical properties of the tumor, such as hypoxia [26] and weak acidity [27], and abnormal metabolic activities, such as the accumulation of adenosine [28] and increased metabolism of L-arginine [29], facilitate the immune escape of tumors.

Traditional immunomodulatory methods

A range of approaches have been developed based on the same goal: to enhance antitumor immunity. Since ITME is an outcome of the parallel occurrence of multiplex protumor mechanisms, combination therapies have the potential to harvest clinical benefits [30]. Immunogenetic cell death (ICD), induced by radiotherapy, phototherapy, and certain chemotherapeutic drugs, such as oxaliplatin, is a type of cell death accompanied by the release of damage-associated molecular patterns (DAMPs) [31]. Tumor cells undergoing ICD will mature dendritic cells (DCs) to cross-present tumor-associated antigens (TAAs) to CD4+ and CD8+ T cells and thus activate an adaptive immune response. In addition, released DAMPs promote phagocytosis and boost the innate immune response. Tumor vaccines or immune adjuvants can also transform cold tumors into hot tumors [32]. Targeted therapies to increase tumor immunogenicity [33], cytokine therapies to activate T cells [34], and oncolytic viruses to release TAAs [35] are potential strategies to propel immunotherapy as well.

However, traditional combination therapies also face the problem of undesired side effects and unsatisfactory efficiency. The nonspecific distribution of therapeutic agents after systemic administration causes damage to healthy organs and reduces the concentration and effects of the drug at the tumor sites. In addition, other therapies in combination with immunotherapy may affect the immune system, which limits the synergetic effects [36]. For instance, lymphodepletion and impact on tertiary lymphoid structures by chemotherapy may hinder the outcome of immunotherapy. The clinical responses of radioimmunotherapy combinations are contradictory since radiation kills tumor cells as well as immune effector cells. Therefore, strategies for targeted drug delivery are necessary to amplify the effects and reduce the toxicity of each component utilized in combination therapy.

NDDSs reversing ITME for enhancing cancer immunotherapy

NDDSs, with higher efficiency and safety than traditional treatments, can deliver immunomodulatory drugs specifically to tumor sites through passive or/and active targeting [37]. They can also be endowed with TME sensitivity to release payloads specifically at tumor sites, which further lessens adverse toxicities [38]. Moreover, multidrug-loaded nanoplatforms provide options for exerting the synergistic effects of combined treatments [39]. In this section, we will mainly introduce the relevant works with the categorization of the strategies to reverse ITME (Fig. 1 and Table 1).

Fig. 1: Nanosized drug delivery systems (NDDSs) modulating the immunosuppressive tumor microenvironment (ITME).
figure 1

Modulating the ITME based on NDDSs through promoting antigen release of tumor cells, maturation of dendritic cells (DCs), activation of cytotoxic T lymphocytes (CTLs), and tumor-killing effects of CTLs. DAMP damage-associated molecular pattern, TAA tumor-associated antigen, ICD immunogenetic cell death, TME tumor microenvironment, TAM tumor-associated macrophage, ROS reactive oxygen species, PD-1/PD-L1 programmed death 1/PD-1 ligand, IDO indoleamine 2,3-dioxygenase.

Table 1 NDDS-based ITME-modulating strategies for improving antitumor immunotherapy.

Combining ICD-inducing therapy with immunotherapy

ICD-inducing treatments provide an immune-activation environment for immunotherapy. Additionally, immunotherapy, including ICB and indoleamine 2,3-dioxygenase (IDO) inhibition, compensates for the upregulation of PD-L1 and IDO caused by IFN-γ, which is secreted by ICD-activated CTLs [40]. NDDSs can deliver targeted toxic ICD-inducing agents, including chemotherapeutic agents, radiotherapy sensitizers, and photosensitizers, to tumor sites, thus minimizing adverse effects and improving therapeutic outcomes [41, 42].

ICD induced by chemotherapy

The combination of chemotherapy and immunotherapy is an encouraging strategy because of the ICD-inducing ability of some chemotherapeutics. To improve the targeting and accessibility of ICD inducers to tumor cells, Li et al. fabricated a bioinspired lipoprotein system containing the legumain-sensitive melittin prodrug, the pH-sensitive phospholipid, and the nitroreductase-sensitive oxaliplatin [43]. After administration, the release of melittin by high-level legumain in tumors promoted the intratumoral permeation of the nanoplatform, and then, the size-enlargement of the nanoplatform after internalization as a response to acidity released oxaliplatin prodrug. Finally, the stimulation of oxaliplatin by nitroreductase and reductive environments induced ICD and elicited antitumor responses (Fig. 2a). Treatment with the nanoplatform increased the proportion of intratumoural CTLs and mature DCs by 1.76-fold and 3.57-fold, respectively, compared with free oxaliplatin treatment, indicating the importance of the delivery strategy. The combination of immune-activating treatment and ICB prolonged the survival of tumor-bearing mice compared with single therapy, demonstrating that preregulating the TME facilitated the effects of immunotherapy. A nanovesicle constructed by fusing artificial liposomes with tumor-derived nanovesicles was utilized for the targeted delivery of doxorubicin, which improved the immunogenicity of tumors and improved the therapeutic efficacy of ICB [44].

Fig. 2: NDDS-based ICD induction for improving immunotherapy.
figure 2

a Schematic illustration of the cancer-accessing tumor-activated size-enlargeable bioinspired lipoprotein system (TA-OBL) to boost antitumor immune responses and synergize with ICB-mediated immunotherapy. b Schematic illustration of the procedures for the evaluation of the activity of sHDL in inducing DC maturation and the proposed mechanism of action. c Treatment-induced extracellular release of ATP and HMGB1 from Hepa1-6 cells treated with different sHDLs. All of the experiments were performed in triplicate, and the data are presented as the mean ± SD (n = 3). *P < 0.05; **P < 0.01; ***P < 0.001. d Schematic illustration of light-inducible nanocargoes (LINC) for improved drug delivery and chemoimmunotherapy by eliciting tumor immunogenicity and overcoming the immunosuppressive tumor microenvironment. e Tumor growth curves in 4T1 tumor-bearing mice after the indicated treatments (n = 6). Data are the mean ± SD. Statistical significance was calculated by one-way ANOVA. a Reproduced from Li et al. [43]. Copyright (2020) John Wiley & Sons. b, c Reproduced from Wang et al. [54]. Copyright (2019) American Chemical Society. d, e Reproduced from Feng et al. [59]. Copyright (2019) John Wiley & Sons.

In addition to sequential delivery, codelivery of ICD inducers and immunotherapeutic agents by NDDSs is also a potential strategy. The tryptophan catabolic enzyme IDO inhibits immune effector cells and promotes immunosuppressive cells [45]. A nanoparticle containing oxaliplatin prodrugs and the IDO inhibitor NLG919 [46] and a biomimetic micelle/monocyte delivery system containing docetaxel, NLG919 and a PD-1/PD-L1 inhibitor [47] were constructed to enhance the efficiency of the drugs and amplify the synergetic effects. These NDDSs provide robust platforms for chemoimmunotherapy to mature DCs and powerfully activate CTLs.

ICD induced by phototherapy

Phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), is capable of inducing ICD by generating reactive oxygen species (ROS) or local hyperthermia, respectively [41]. Given the high expression of matrix metalloproteinase 2 (MMP-2) in the TME, Wang and coworkers encapsulated an anti-PD-L1 antibody and a photosensitizer into MMP-2-responsive nanoparticles to precisely release drugs at the tumor site. Photosensitizer-mediated PDT under a near-infrared (NIR) laser sensitized the tumors to ICB [48]. Consequently, the tumor inhibition rate of mice treated with the nanoplatform and irradiation was 73.2%, which was only 39.8% of the free antibody-treated group. Similarly, a pH-sensitive nanoplatform loaded with a photosensitizer and a PD-L1 siRNA was constructed to overcome the immunological tolerance of tumors [49]. TME-activated nanoplatforms codelivering photosensitizers and IDO inhibitors [50, 51] or Toll-like receptor 3 (TLR3) agonists [

References

  1. Khalil DN, Smith EL, Brentjens RJ, Wolchok JD. The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol. 2016;13:273–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sanmamed MF, Chen L. A paradigm shift in cancer immunotherapy: from enhancement to normalization. Cell. 2018;175:313–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat Rev Cancer. 2016;16:275–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. June CH, Warshauer JT, Bluestone JA. Is autoimmunity the achilles’ heel of cancer immunotherapy? Nat Med. 2017;23:540–7.

    Article  CAS  PubMed  Google Scholar 

  5. Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378:158–68.

    Article  CAS  PubMed  Google Scholar 

  6. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168:707–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition. N Engl J Med. 2017;377:2500–1.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Tormoen GW, Crittenden MR, Gough MJ. Role of the immunosuppressive microenvironment in immunotherapy. Adv Radiat Oncol. 2018;3:520–6.

    Article  PubMed  PubMed Central  Google Scholar 

  9. O’Donnell JS, Teng MWL, Smyth MJ. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat Rev Clin Oncol. 2019;16:151–67.

    Article  PubMed  Google Scholar 

  10. Phuengkham H, Ren L, Shin IW, Lim YT. Nanoengineered immune niches for reprogramming the immunosuppressive tumor microenvironment and enhancing cancer immunotherapy. Adv Mater. 2019;31:e1803322.

    Article  PubMed  Google Scholar 

  11. Liu X, Wang D, Zhang P, Li Y. Recent advances in nanosized drug delivery systems for overcoming the barriers to anti-PD immunotherapy of cancer. Nano Today. 2019;29:100801.

    Article  CAS  Google Scholar 

  12. Zhang P, Zhai Y, Cai Y, Zhao Y, Li Y. Nanomedicine-based immunotherapy for the treatment of cancer metastasis. Adv Mater. 2019;31:e1904156.

    Article  PubMed  Google Scholar 

  13. Goldberg MS. Improving cancer immunotherapy through nanotechnology. Nat Rev Cancer. 2019;19:587–602.

    Article  CAS  PubMed  Google Scholar 

  14. Nam J, Son S, Park KS, Zou W, Shea LD, Moon JJ. Cancer nanomedicine for combination cancer immunotherapy. Nat Rev Mater. 2019;4:398–414.

    Article  Google Scholar 

  15. Qi FL, Wang MF, Li BZ, Lu ZF, Nie GJ, Li SP. Reversal of the immunosuppressive tumor microenvironment by nanoparticle-based activation of immune-associated cells. Acta Pharmacol Sin. 2020;41:895–901.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Sun B, Hyun H, Li LT, Wang AZ. Harnessing nanomedicine to overcome the immunosuppressive tumor microenvironment. Acta Pharmacol Sin. 2020;41:970–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Vitale I, Shema E, Loi S, Galluzzi L. Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat Med. 2021;27:212–24.

    Article  CAS  PubMed  Google Scholar 

  18. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24:541–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov. 2019;18:197–218.

    Article  CAS  PubMed  Google Scholar 

  20. Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, et al. Cold tumors: a therapeutic challenge for immunotherapy. Front Immunol. 2019;10:168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Samstein RM, Lee C-H, Shoushtari AN, Hellmann MD, Shen R, Janjigian YY, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet. 2019;51:202–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Westcott PMK, Sacks NJ, Schenkel JM, Ely ZA, Smith O, Hauck H, et al. Low neoantigen expression and poor T-cell priming underlie early immune escape in colorectal cancer. Nat Cancer. 2021;2:1071–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li L, Yu R, Cai T, Chen Z, Lan M, Zou T, et al. Effects of immune cells and cytokines on inflammation and immunosuppression in the tumor microenvironment. Int Immunopharmacol. 2020;88:106939.

    Article  CAS  PubMed  Google Scholar 

  24. Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, et al. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther. 2021;6:153.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mao X, Xu J, Wang W, Liang C, Hua J, Liu J, et al. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol Cancer. 2021;20:131.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. You L, Wu W, Wang X, Fang L, Adam V, Nepovimova E, et al. The role of hypoxia-inducible factor 1 in tumor immune evasion. Med Res Rev. 2021;41:1622–43.

    Article  CAS  PubMed  Google Scholar 

  27. Wang JX, Choi SYC, Niu X, Kang N, Xue H, Killam J, et al. Lactic acid and an acidic tumor microenvironment suppress anticancer immunity. Int J Mol Sci. 2020;21:8363.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Leone RD, Emens LA. Targeting adenosine for cancer immunotherapy. J Immunother Cancer. 2018;6:57.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, et al. L-arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell. 2016;167:1–14.

    Article  Google Scholar 

  30. Xue G, Wang Z, Zheng N, Fang J, Mao C, Li X, et al. Elimination of acquired resistance to PD-1 blockade via the concurrent depletion of tumour cells and immunosuppressive cells. Nat Biomed Eng. 2021;5:1306–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer. 2012;12:860–75.

    Article  CAS  PubMed  Google Scholar 

  32. Paston SJ, Brentville VA, Symonds P, Durrant LG. Cancer vaccines, adjuvants, and delivery systems. Front Immunol. 2021;12:627932.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. de Charette M, Marabelle A, Houot R. Turning tumour cells into antigen presenting cells: the next step to improve cancer immunotherapy? Eur J Cancer. 2016;68:134–47.

    Article  PubMed  Google Scholar 

  34. Berraondo P, Sanmamed MF, Ochoa MC, Etxeberria I, Aznar MA, Pérez-Gracia JL, et al. Cytokines in clinical cancer immunotherapy. Br J Cancer. 2019;120:6–15.

    Article  CAS  PubMed  Google Scholar 

  35. Kaufman HL, Kohlhapp FJ, Zloza A. Oncolytic viruses: a new class of immunotherapy drugs. Nat Rev Drug Discov. 2015;14:642–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhu S, Zhang T, Zheng L, Liu H, Song W, Liu D, et al. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol. 2021;14:156.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yin W, Li Y, Gu Y, Luo M. Nanoengineered targeting strategy for cancer immunotherapy. Acta Pharmacol Sin. 2020;41:902–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang Z, Wang H, Tan T, Li J, Wang Z, Li Y. Rational design of nanoparticles with deep tumor penetration for effective treatment of tumor metastasis. Adv Funct Mater. 2018;28:1801840.

    Article  Google Scholar 

  39. Chen J, Zhu Y, Wu C, Shi J. Nanoplatform-based cascade engineering for cancer therapy. Chem Soc Rev. 2020;49:9057–94.

    Article  CAS  PubMed  Google Scholar 

  40. Castro F, Cardoso AP, Gonçalves RM, Serre K, Oliveira MJ. Interferon-gamma at the crossroads of tumor immune surveillance or evasion. Front Immunol. 2018;9:847.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Yan W, Lang T, Qi X, Li Y. Engineering immunogenic cell death with nanosized drug delivery systems improving cancer immunotherapy. Curr Opin Biotechnol. 2020;66:36–43.

    Article  CAS  PubMed  Google Scholar 

  42. Zhou L, Zhang P, Wang H, Wang D, Li Y. Smart nanosized drug delivery systems inducing immunogenic cell death for combination with cancer immunotherapy. Acc Chem Res. 2020;53:1761–72.

    Article  CAS  PubMed  Google Scholar 

  43. Li J, Wang H, Wang Y, Gong X, Xu X, Sha X, et al. Tumor-activated size-enlargeable bioinspired lipoproteins access cancer cells in tumor to elicit anti-tumor immune responses. Adv Mater. 2020;32:e2002380.

    Article  PubMed  Google Scholar 

  44. Hu M, Zhang J, Kong L, Yu Y, Hu Q, Yang T, et al. Immunogenic hybrid nanovesicles of liposomes and tumor-derived nanovesicles for cancer immunochemotherapy. ACS Nano. 2021;15:3123–38.

    Article  CAS  PubMed  Google Scholar 

  45. Löb S, Königsrainer 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. 2009;9:445–52.

    Article  PubMed  Google Scholar 

  46. Feng B, Zhou F, Hou B, Wang D, Wang T, Fu Y, et al. Binary cooperative prodrug nanoparticles improve immunotherapy by synergistically modulating immune tumor microenvironment. Adv Mater. 2018;30:1803001.

    Article  Google Scholar 

  47. Lang T, Zheng Z, Huang X, Liu Y, Zhai Y, Zhang P, et al. Ternary regulation of tumor microenvironment by heparanase-sensitive micelle-loaded monocytes improves chemo-immunotherapy of metastatic breast cancer. Adv Funct Mater. 2021;31:2007402.

    Article  CAS  Google Scholar 

  48. Wang D, Wang T, Yu H, Feng B, Zhou L, Zhou F, et al. Engineering nanoparticles to locally activate T cells in the tumor microenvironment. Sci Immunol. 2019;4:eaau6584.

    Article  CAS  PubMed  Google Scholar 

  49. Wang D, Wang T, Liu J, Yu H, Jiao S, Feng B, et al. Acid-activatable versatile micelleplexes for PD-L1 blockade-enhanced cancer photodynamic immunotherapy. Nano Lett. 2016;16:5503–13.

    Article  CAS  PubMed  Google Scholar 

  50. Gao A, Chen B, Gao J, Zhou F, Saeed M, Hou B, et al. Sheddable prodrug vesicles combating adaptive immune resistance for improved photodynamic immunotherapy of cancer. Nano Lett. 2020;20:353–62.

    Article  CAS  PubMed  Google Scholar 

  51. Hou B, Zhou L, Wang H, Saeed M, Wang D, Xu Z, et al. Engineering stimuli-activatable boolean logic prodrug nanoparticles for combination cancer immunotherapy. Adv Mater. 2020;32:e1907210.

    Article  PubMed  Google Scholar 

  52. Fang L, Zhao Z, Wang J, **ao P, Sun X, Ding Y, et al. Light-controllable charge-reversal nanoparticles with polyinosinic-polycytidylic acid for enhancing immunotherapy of triple negative breast cancer. Acta Pharmacol Sin B 2022;12:353–63.

    Article  CAS  Google Scholar 

  53. Wang Z, Gong X, Li J, Wang H, Xu X, Li Y, et al. Oxygen-delivering polyfluorocarbon nanovehicles improve tumor oxygenation and potentiate photodynamic-mediated antitumor immunity. ACS Nano. 2021;15:5405–19.

    Article  CAS  PubMed  Google Scholar 

  54. Wang J, Meng J, Ran W, Lee RJ, Teng L, Zhang P, et al. Hepatocellular carcinoma growth retardation and pd-1 blockade therapy potentiation with synthetic high-density lipoprotein. Nano Lett. 2019;19:5266–76.

    Article  CAS  PubMed  Google Scholar 

  55. Zhou T, Liang X, Wang P, Hu Y, Qi Y, ** Y, et al. A hepatocellular carcinoma targeting nanostrategy with hypoxia-ameliorating and photothermal abilities that, combined with immunotherapy, inhibits metastasis and recurrence. ACS Nano. 2020;14:12679–96.

    Article  CAS  PubMed  Google Scholar 

  56. Formenti SC, Demaria S. Systemic effects of local radiotherapy. Lancet Oncol. 2009;10:718–26.

    Article  PubMed  PubMed Central  Google Scholar 

  57. ** J, Zhao Q. Engineering nanoparticles to reprogram radiotherapy and immunotherapy: Recent advances and future challenges. J Nanobiotechnology. 2020;18:75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wang Y, Chen J, Duan R, Gu R, Wang W, Wu J, et al. High-Z-sensitized radiotherapy synergizes with the intervention of the pentose phosphate pathway for in situ tumor vaccination. Adv Mater. 2022;34:2109726.

    Article  CAS  Google Scholar 

  59. Feng B, Hou B, Xu Z, Saeed M, Yu H, Li Y. Self-amplified drug delivery with light-inducible nanocargoes to enhance cancer immunotherapy. Adv Mater. 2019;31:e1902960.

    Article  PubMed  Google Scholar 

  60. Zhou F, Feng B, Yu H, Wang D, Wang T, Ma Y, et al. Tumor microenvironment-activatable prodrug vesicles for nanoenabled cancer chemoimmunotherapy combining immunogenic cell death induction and CD47 blockade. Adv Mater. 2019;31:e1805888.

    Article  PubMed  Google Scholar 

  61. Morse MA, Gwin WR, Mitchell DA. Vaccine therapies for cancer: then and now. Target Oncol. 2021;16:121–52.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Saxena M, van der Burg SH, Melief CJM, Bhardwaj N. Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21:360–78.

    Article  CAS  PubMed  Google Scholar 

  63. Das A, Ali N. Nanovaccine: an emerging strategy. Expert Rev Vaccines. 2021;20:1273–90.

    Article  CAS  PubMed  Google Scholar 

  64. Wang T, Wang D, Yu H, Feng B, Zhou F, Zhang H, et al. A cancer vaccine-mediated postoperative immunotherapy for recurrent and metastatic tumors. Nat Commun. 2018;9:1532.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Fang L, Zhao Z, Wang J, Zhang P, Ding Y, Jiang Y, et al. Engineering autologous tumor cell vaccine to locally mobilize antitumor immunity in tumor surgical bed. Sci Adv. 2020;6:eaba4024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Zhou L, Hou B, Wang D, Sun F, Song R, Shao Q, et al. Engineering polymeric prodrug nanoplatform for vaccination immunotherapy of cancer. Nano Lett. 2020;20:4393–402.

    Article  CAS  PubMed  Google Scholar 

  67. Jiang Y, Krishnan N, Zhou J, Chekuri S, Wei X, Kroll AV, et al. Engineered cell-membrane-coated nanoparticles directly present tumor antigens to promote anticancer immunity. Adv Mater. 2020;32:e2001808.

    Article  PubMed  PubMed Central  Google Scholar 

  68. **ao P, Wang J, Zhao Z, Liu X, Sun X, Wang D, et al. Engineering nanoscale artificial antigen-presenting cells by metabolic dendritic cell labeling to potentiate cancer immunotherapy. Nano Lett. 2021;21:2094–103.

    Article  CAS  PubMed  Google Scholar 

  69. **ao P, Wang J, Fang L, Zhao Z, Sun X, Liu X, et al. Nanovaccine-mediated cell selective delivery of neoantigens potentiating adoptive dendritic cell transfer for personalized immunization. Adv Funct Mater. 2021;31:2104068.

    Article  CAS  Google Scholar 

  70. Liu C, Liu X, **ang X, Pang X, Chen S, Zhang Y, et al. A nanovaccine for antigen self-presentation and immunosuppression reversal as a personalized cancer immunotherapy strategy. Nat Nanotechnol. 2022;17:531–40.

    Article  CAS  PubMed  Google Scholar 

  71. Wang J, Zheng C, Zhai Y, Cai Y, Lee RJ, **ng J, et al. High-density lipoprotein modulates tumor-associated macrophage for chemoimmunotherapy of hepatocellular carcinoma. Nano Today. 2021;37:101064.

    Article  CAS  Google Scholar 

  72. Xu X, Gong X, Wang Y, Li J, Wang H, Wang J, et al. Reprogramming tumor associated macrophages toward M1 phenotypes with nanomedicine for anticancer immunotherapy. Adv Ther. 2020;3:1900181.

    Article  Google Scholar 

  73. Wei Z, Zhang X, Yong T, Bie N, Zhan G, Li X, et al. Boosting anti-PD-1 therapy with metformin-loaded macrophage-derived microparticles. Nat Commun. 2021;12:440.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Rosenberg SA. IL-2: The first effective immunotherapy for human cancer. J Immunol. 2014;192:5451–8.

    Article  CAS  PubMed  Google Scholar 

  75. Propper DJ, Balkwill FR. Harnessing cytokines and chemokines for cancer therapy. Nat Rev Clin Oncol. 2022;19:237–53.

    Article  CAS  PubMed  Google Scholar 

  76. Zhai Y, Wang J, Lang T, Kong Y, Rong R, Cai Y, et al. T lymphocyte membrane-decorated epigenetic nanoinducer of interferons for cancer immunotherapy. Nat Nanotechnol. 2021;16:1271–80.

    Article  CAS  PubMed  Google Scholar 

  77. Wu T, Qiao Q, Qin X, Zhang D, Zhang Z. Immunostimulatory cytokine and doxorubicin co-loaded nanovesicles for cancer immunochemotherapy. Nanomedicine. 2019;18:66–77.

    Article  CAS  PubMed  Google Scholar 

  78. Hu Q, Shang L, Wang M, Tu K, Hu M, Yu Y, et al. Co-delivery of paclitaxel and interleukin-12 regulating tumor microenvironment for cancer immunochemotherapy. Adv Health Mater. 2020;9:e1901858.

    Article  Google Scholar 

  79. Feng B, Niu Z, Hou B, Zhou L, Li Y, Yu H. Enhancing triple negative breast cancer immunotherapy by ICG-templated self-assembly of paclitaxel nanoparticles. Adv Funct Mater. 2020;30:1906605.

    Article  CAS  Google Scholar 

  80. Wang H, Li J, Wang Z, Wang Y, Xu X, Gong X, et al. Tumor-permeated bioinspired theranostic nanovehicle remodels tumor immunosuppression for cancer therapy. Biomaterials. 2021;269:120609.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

National Natural Science Foundation of China (81871471, 31930066, 32130058, and 32171315), Natural Science Foundation of Shanghai (19ZR1479900), Science and Technology Commission of Shanghai Municipality (19431900800), International Partnership Program of CAS (153631KYSB20190013), Natural Science Foundation of Shandong (ZR2019ZD25), Special Research Assistant Project of CAS, China Postdoctoral Science Foundation (2020M681428), and Shanghai Postdoctoral Excellence Program (2020495) are gratefully acknowledged for financial support.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Qi Yin or Ya-** Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Rights and permissions

Springer Nature or its licensor 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

Yan, Wl., Lang, Tq., Yuan, Wh. et al. Nanosized drug delivery systems modulate the immunosuppressive microenvironment to improve cancer immunotherapy. Acta Pharmacol Sin 43, 3045–3054 (2022). https://doi.org/10.1038/s41401-022-00976-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41401-022-00976-6

  • Springer Nature Singapore Pte Ltd.

Keywords