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

Emerging advances in nanomedicine for breast cancer immunotherapy: opportunities and challenges

    Mohammad Zaki Ahmad

    *Author for correspondence:

    E-mail Address: mzahmad@nu.edu.sa

    Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia

    ,
    Ali S Alasiri

    Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia

    ,
    Mohammed Yahia Alasmary

    Medical Department, College of Medicine, Najran University, Najran, 11001, Kingdom of Saudi Arabia

    ,
    MM Abdullah

    Advanced Materials & Nano-Research Centre, Department of Physics, Faculty of Science & Arts, Najran University, Najran, 11001, Kingdom Saudi Arabia

    ,
    Javed Ahmad

    Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Najran, 11001, Kingdom of Saudi Arabia

    ,
    Basel A Abdel Wahab

    Department of Pharmacology, College of Pharmacy, Najran University, Najran, 11001, Kingdom of Saudi Arabia

    Department of Pharmacology, College of Medicine, Assiut University, Assiut, 71515, Egypt

    ,
    Saif Aboud M Alqahtani

    Internal Medicine Department, College of Medicine, King Khalid University, Abha, 61421, Kingdom of Saudi Arabia

    ,
    Kalyani Pathak

    Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India

    ,
    Gulam Mustafa

    College of Pharmacy, Shaqra University, Ad-Dawadmi Riyadh, Kingdom of Saudi Arabia

    ,
    Mohammad Ahmad Khan

    Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India

    ,
    Riya Saikia

    Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India

    &
    Urvashee Gogoi

    Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, 786004, Assam, India

    Published Online:https://doi.org/10.2217/imt-2021-0348

    Breast cancer is one of the most common causes of cancer-related morbidity and mortality in women worldwide. Early diagnosis and an appropriate therapeutic approach for all cancers are climacterics for a favorable prognosis. Targeting the immune system in breast cancer is already a clinical reality with notable successes, specifically with checkpoint blockade antibodies and chimeric antigen receptor T-cell therapy. However, there have been inevitable setbacks in the clinical application of cancer immunotherapy, including inadequate immune responses due to insufficient delivery of immunostimulants to immune cells and uncontrolled immune system modulation. Rapid advancements and new evidence have suggested that nanomedicine-based immunotherapy may be a viable option for treating breast cancer.

    Plain language summary

    Cancer that begins in the breast is referred to as breast cancer. It may originate in either one or both breasts. It is one of the main causes of cancer-related death among women worldwide. Cancer immunotherapy is a game-changing treatment that improves the ability of the host defense system to spot and eliminate cancer cells with pinpoint accuracy. Cancer immunotherapy, also referred to as immuno-oncology, is a type of treatment option for breast cancer that uses the body’s natural defense system to prevent, regulate and eliminate breast cancer. Immunotherapy is used to enhance or alter the functioning of the immune system so that it can locate and destroy cancer cells. Knowing how immunotherapy works and what to anticipate can often offer peace of mind to the patient who can then make informed decisions about care, especially if immunotherapy is part of the treatment plan for a particular patient.

    Tweetable abstract

    Breast cancer is the most common cancer in women. Rapid advancements and new evidence have suggested that #nanomedicine-based immunotherapy may be a viable option for treating #breastcancer

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

    Reference

    • 1. Clark BZ, Onisko A, Assylbekova B et al. Breast cancer global tumor biomarkers: a quality assurance study of intratumoral heterogeneity. Mod. Pathol. 32(3), 354–366 (2019).
    • 2. Mahvi DA, Liu R, Grinstaff MW et al. Local cancer recurrence: the realities, challenges, and opportunities for new therapies. CA Cancer J. Clin. 68(6), 488–505 (2018).
    • 3. Parada H Jr, Sun X, Tse CK et al. Lifestyle patterns and survival following breast cancer in the Carolina Breast Cancer Study. Epidemiology 30(1), 83–92 (2019).
    • 4. Cain EH, Saha A, Harowicz MR et al. Multivariate machine learning models for prediction of pathologic response to neoadjuvant therapy in breast cancer using MRI features: a study using an independent validation set. Breast Cancer Res. Treat. 173(2), 455–463 (2019).
    • 5. Rizwanullah M, Ahmad MZ, Ghoneim MM et al. Receptor-mediated targeted delivery of surface-modifiednanomedicine in breast cancer: recent update and challenges. Pharmaceutics 13(12), 2039 (2021). •• Excellent review article summarizing the receptor mediated delivery of nanomedicine in breast cancer.
    • 6. Inic Z, Zegarac M, Inic M et al. Difference between luminal a and luminal b subtypes according to Ki-67, tumor size, and progesterone receptor negativity providing prognostic information. Clin. Med. Insights Oncol. 8, 107–111 (2014).
    • 7. Fragomeni SM, Sciallis A, Jeruss JS. Molecular subtypes and local-regional control of breast cancer. Surg. Oncol. Clin. N Am 27(1), 95–120 (2018).
    • 8. Ahmad MZ, Ahmad J, Alasmary MY et al. Emerging advances in cationic liposomal cancer nanovaccines: opportunities and challenges. Immunotherapy 13(6), 491–507 (2021). •• Review article dealing with the cationic lipsomal cancer nanovaccines.
    • 9. Ahmad MZ, Ahmad J, Haque A et al. Emerging advances in synthetic cancer nano-vaccines: opportunities and challenges. Expert Rev. Vaccines 19(11), 1053–1071 (2020).
    • 10. Coulie PG, Van Den Eynde BJ, Van Der Bruggen P, Boon T. Tumour antigens recognized by T lymphocytes: at the core of cancer immunotherapy. Nat. Rev. Cancer 14(2), 135–146 (2014).
    • 11. Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature 541(7637), 321–330 (2017).
    • 12. Hewitt EW. The MHC class I antigen presentation pathway: strategies for viral immune evasion. Immunology 110(2), 163–169 (2003).
    • 13. Raval RR, Sharabi AB, Walker AJ et al. Tumor immunology and cancer immunotherapy: summary of the 2013 SITC primer. J. Immunother. Cancer 2, 14 (2014).
    • 14. Tan TJ, Chan JJ, Kamis S, Dent RA. What is the role of immunotherapy in breast cancer? Chinese Clin. Oncol. 7(2), 13 (2018).
    • 15. Shao K, Singha S, Clemente-Casares X et al. Nanoparticle-based immunotherapy for cancer. ACS Nano 9(1), 16–30 (2015).
    • 16. De Melo Gagliato D, Buzaid AC, Perez-Garcia J, Cortes J. Immunotherapy in breast cancer: current practice and clinical challenges. BioDrugs 34(5), 611–623 (2020).
    • 17. Goff SL, Danforth DN. The role of immune cells in breast tissue and immunotherapy for the treatment of breast cancer. Clin. Breast Cancer 21(1), e63–e73 (2021).
    • 18. Mina LA, Lim S, Bahadur SW, Firoz AT. Immunotherapy for the treatment of breast cancer: emerging new data. Breast Cancer 11, 321–328 (2019).
    • 19. Underwood JC. Lymphoreticular infiltration in human tumours: prognostic and biological implications: a review. Br. J. Cancer 30(6), 538–548 (1974).
    • 20. Aaltomaa S, Lipponen P, Eskelinen M et al. Lymphocyte infiltrates as a prognostic variable in female breast cancer. Eur. J. Cancer 28a(4–5), 859–864 (1992).
    • 21. Chin Y, Janseens J, Vandepitte J et al. Phenotypic analysis of tumor-infiltrating lymphocytes from human breast cancer. Anticancer Res. 12(5), 1463–1466 (1992).
    • 22. Soliman H, Khalil F, Antonia S. PD-L1 expression is increased in a subset of basal type breast cancer cells. PLOS ONE 9(2), e88557 (2014).
    • 23. Sabatier R, Finetti P, Mamessier E et al. Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget 6(7), 5449–5464 (2015).
    • 24. Sharpe AH, Wherry EJ, Ahmed R, Freeman GJ. The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nat. Immunol. 8(3), 239–245 (2007).
    • 25. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am. J. Cancer Res. 10(3), 727–742 (2020).
    • 26. Yu LY, Tang J, Zhang CM et al. New immunotherapy strategies in breast cancer. Int. J. Environ. Res. Public Health 14(1), 68 (2017).
    • 27. Ohaegbulam KC, Assal A, Lazar-Molnar E, Yao Y, Zang X. Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway. Trends Mol. Med. 21(1), 24–33 (2015).
    • 28. Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39(1), 1–10 (2013).
    • 29. Shi Y, Lammers T. Combining nanomedicine and immunotherapy. Acc. Chem. Res. 52(6), 1543–1554 (2019).
    • 30. Okazaki T, Chikuma S, Iwai Y et al. A rheostat for immune responses: the unique properties of PD-1 and their advantages for clinical application. Nat. Immunol. 14(12), 1212–1218 (2013).
    • 31. Basu A, Ramamoorthi G, Jia Y et al. Immunotherapy in breast cancer: current status and future directions. Adv. Cancer Res. 143, 295–349 (2019).
    • 32. Li Y, Miao W, He D et al. Recent progress on immunotherapy for breast cancer: tumor microenvironment, nanotechnology and more. Front. Bioeng. Biotechnol. 9, 680315 (2021).
    • 33. Azvolinsky A. What is tumor mutational burden? Cancer Today (2020). www.cancertodaymag.org/Pages/cancer-talk/What-Is-Tumor-Mutational-Burden.aspx
    • 34. Mariathasan S, Turley SJ, Nickles D et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 554(7693), 544–548 (2018).
    • 35. Shao C, Li G, Huang L et al. Prevalence of high tumor mutational burden and association with survival in patients with less common solid tumors. JAMA Netw. Open 3(10), e2025109 (2020).
    • 36. Yarchoan M, Albacker LA, Hopkins AC et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight 4(6), e126908 (2019).
    • 37. Rizvi NA, Cho BC, Reinmuth N et al. Durvalumab with or without tremelimumab vs platinum-based chemotherapy as first-line treatment for metastatic non-small cell lung cancer: MYSTIC. Ann. Oncol. 29, x40–x41 (2018).
    • 38. Danaher P, Warren S, Lu R et al. Pan-cancer adaptive immune resistance as defined by the Tumor Inflammation Signature (TIS): results from The Cancer Genome Atlas (TCGA). J. Immunother. Cancer 6(1), 63 (2018).
    • 39. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science 348(6230), 69–74 (2015).
    • 40. Alexandrov LB, Nik-Zainal S, Wedge DC et al. Signatures of mutational processes in human cancer. Nature 500(7463), 415–421 (2013).
    • 41. National Cancer Institute. Definition of TIL. NCI Dictionary of Cancer Terms (2022). www.cancer.gov/publications/dictionaries/cancer-terms/def/til
    • 42. Basu A, Ramamoorthi G, Jia Y et al. Chapter Six: Immunotherapy in breast cancer: current status and future directions. In: Advances in Cancer Research Wang X-YFisher PB (Eds). Academic Press, MA, USA, 295–349 (2019).
    • 43. Ruan M, Tian T, Rao J et al. Predictive value of tumor-infiltrating lymphocytes to pathological complete response in neoadjuvant treated triple-negative breast cancers. Diagn. Pathol. 13(1), 66 (2018).
    • 44. Ravelli A, Roviello G, Cretella D et al. Tumor-infiltrating lymphocytes and breast cancer: beyond the prognostic and predictive utility. Tumour Biol. 39(4), 1010428317695023 (2017).
    • 45. Stanton SE, Adams S, Disis ML. Variation in the Incidence and Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes: A Systematic Review. JAMA Oncol. 2(10), 1354–1360 (2016).
    • 46. Denkert C, Von Minckwitz G, Darb-Esfahani S et al. Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 19(1), 40–50 (2018).
    • 47. Loi S, Michiels S, Salgado R et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: results from the FinHER trial. Ann. Oncol. 25(8), 1544–1550 (2014).
    • 48. Salgado R, Denkert C, Campbell C et al. Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast cancer treated with lapatinib and trastuzumab: a secondary analysis of the NeoALTTO Trial. JAMA Oncol. 1(4), 448–454 (2015).
    • 49. Perez EA, Ballman KV, Tenner KS et al. Association of stromal tumor-infiltrating lymphocytes with recurrence-free survival in the N9831 adjuvant trial in patients with early-stage HER2-positive breast cancer. JAMA Oncol. 2(1), 56–64 (2016).
    • 50. Luen SJ, Salgado R, Fox S et al. Tumour-infiltrating lymphocytes in advanced HER2-positive breast cancer treated with pertuzumab or placebo in addition to trastuzumab and docetaxel: a retrospective analysis of the CLEOPATRA study. Lancet Oncol. 18(1), 52–62 (2017).
    • 51. Gentles AJ, Newman AM, Liu CL et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21(8), 938–945 (2015).
    • 52. Li W, Wei H, Li H, Gao J et al. Cancer nanoimmunotherapy using advanced pharmaceutical nanotechnology. Nanomedicine 9(16), 2587–2605 (2014).
    • 53. Dunn GP, Bruce AT, Ikeda H et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol. 3(11), 991–998 (2002).
    • 54. Hashemzadeh N, Dolatkhah M, Adibkia K et al. Recent advances in breast cancer immunotherapy: the promising impact of nanomedicines. Life Sci. 271, 119110 (2021).
    • 55. Matthaiou EI, Barar J, Sandaltzopoulos R et al. Shikonin-loaded antibody-armed nanoparticles for targeted therapy of ovarian cancer. Int. J. Nanomedicine 9, 1855–1870 (2014).
    • 56. Hashemzadeh N, Dolatkhah M, Aghanejad A et al. Folate receptor-mediated delivery of 1-MDT-loaded mesoporous silica magnetic nanoparticles to target breast cancer cells. Nanomedicine 16(24), 2137–2154 (2021).
    • 57. Goldberg MS. Improving cancer immunotherapy through nanotechnology. Nat. Rev. Cancer 19(10), 587–602 (2019).
    • 58. Yu HJ, De Geest BG. Nanomedicine and cancer immunotherapy. Acta Pharmacol. Sinica 41(7), 879–880 (2020).
    • 59. Ahmad MZ, Akhter S, Mohsin N et al. Transformation of curcumin from food additive to multifunctional medicine: nanotechnology bridging the gap. Curr. Drug Discov. Technol. 11(3), 197–213 (2014).
    • 60. Ahmad MZ, Akhter S, Rahman Z et al. Nanometric gold in cancer nanotechnology: current status and future prospect. J. Pharm. Pharmacol. 65(5), 634–651 (2013).
    • 61. Ahmad MZ, Alkahtani SA, Akhter S et al. Progress in nanotechnology-based drug carrier in designing of curcumin nanomedicines for cancer therapy: current state-of-the-art. J. Drug Target. 24(4), 273–293 (2016).
    • 62. Ahmad MZ, Rizwanullah M, Ahmad J et al. Progress in nanomedicine-based drug delivery in designing of chitosan nanoparticles for cancer therapy. Int. J. Polym. Mater. Polym. Biomater. 71(8), 602–623 (2021).
    • 63. Akhter S, Ahmad Z, Singh A et al. Cancer targeted metallic nanoparticle: targeting overview, recent advancement and toxicity concern. Curr. Pharm. Des. 17(18), 1834–1850 (2011).
    • 64. Algahtani MS, Ahmad MZ, Nourein IH, Ahmad J. Co-delivery of imiquimod and curcumin by nanoemugel for improved topical delivery and reduced psoriasis-like skin lesions. Biomolecules 10(7), 968 (2020).
    • 65. Algahtani MS, Ahmad MZ, Shaikh IA et al. Thymoquinone loaded topical nanoemulgel for wound healing: formulation design and in-vivo evaluation. Molecules 26(13), 3863 (2021).
    • 66. Algahtani MS, Ahmad MZ, Ahmad J. Nanoemulsion loaded polymeric hydrogel for topical delivery of curcumin in psoriasis. J. Drug Deliv. Sci. Technol. 59, 101847 (2020).
    • 67. Fernandez-Fernandez A, Manchanda R, Mcgoron AJ. Theranostic applications of nanomaterials in cancer: drug delivery, image-guided therapy, and multifunctional platforms. Appl. Biochem. Biotechnol. 165(7–8), 1628–1651 (2011).
    • 68. Ahmad MZ, Akhter S, Jain GK et al. Metallic nanoparticles: technology overview & drug delivery applications in oncology. Expert Opin. Drug Deliv. 7(8), 927–942 (2010).
    • 69. Gong N, Zhang Y, Zhang Z, Li X, Liang XJ. Functional nanomaterials optimized to circumvent tumor immunological tolerance. Adv. Functional Materials 29(3), 1806087 (2019).
    • 70. Huang P, Wang X, Liang X et al. Nano-, micro-, and macroscale drug delivery systems for cancer immunotherapy. Acta Biomater 85, 1–26 (2019).
    • 71. Park YM, Lee SJ, Kim YS et al. Nanoparticle-based vaccine delivery for cancer immunotherapy. Immune Netw. 13(5), 177–183 (2013).
    • 72. Fu B, Huang X, Deng J et al. Application of multifunctional nanomaterials in cancer vaccines (Review). Oncol. Rep. 39(3), 893–900 (2018).
    • 73. Yu X, Dai Y, Zhao Y et al. Melittin-lipid nanoparticles target to lymph nodes and elicit a systemic anti-tumor immune response. Nat. Commun. 11(1), 1110 (2020).
    • 74. Kumar S, Anselmo AC, Banerjee A et al. Shape and size-dependent immune response to antigen-carrying nanoparticles. J. Control ReL. 220(Pt A), 141–148 (2015).
    • 75. Grimaldi AM, Incoronato M, Salvatore M, Soricelli A. Nanoparticle-based strategies for cancer immunotherapy and immunodiagnostics. Nanomedicine 12(19), 2349–2365 (2017). • Nanomedicine-based cancer immunotherapy and immunodiagnostic.
    • 76. Vangijzegem T, Stanicki D, Laurent S. Magnetic iron oxide nanoparticles for drug delivery: applications and characteristics. Expert Opin. Drug Deliv. 16(1), 69–78 (2019).
    • 77. Ahmad MZ, Ahmad J, Zafar S et al. Omega-3 fatty acids as adjunctive therapeutics: prospective of nanoparticles in its formulation development. Ther. Deliv. 11(1), 851–868 (2020).
    • 78. Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: from basics to clinical immunization. J. Control Rel. 303, 130–150 (2019). •• Nanoliposomes as delivery systems for cancer immunotherapy
    • 79. Allison AC, Gregoriadis G. Liposomes as immunological adjuvants. In: Lymphocytes, Macrophages, and Cancer. Mathé GFlorentin ISimmler M-C (Eds). Springer Berlin-Heidelberg, Berlin/Heidelberg, Germany, 58–64 (1976).
    • 80. Beiu C, Giurcaneanu C, Grumezescu AM et al. Nanosystems for improved targeted therapies in melanoma. J. Clin. Med. 9(2), 318 (2020).
    • 81. Perez SA, Anastasopoulou EA, Tzonis P et al. AE37 peptide vaccination in prostate cancer: a 4-year immunological assessment updates on a Phase I trial. Cancer Immunol. Immunother. 62(10), 1599–1608 (2013).
    • 82. Bode C, Zhao G, Steinhagen F et al. CpG DNA as a vaccine adjuvant. Expert Rev. Vaccines 10(4), 499–511 (2011).
    • 83. Barati N, Nikpoor AR, Razazan A et al. Nanoliposomes carrying HER2/neu-derived peptide AE36 with CpG-ODN exhibit therapeutic and prophylactic activities in a mice TUBO model of breast cancer. Immunol. Lett. 190, 108–117 (2017).
    • 84. Barati N, Nikpoor AR, Mosaffa F et al. AE36 HER2/neu-derived peptide linked to positively charged liposomes with CpG-ODN as an effective therapeutic and prophylactic vaccine for breast cancer. J. Drug Deliv. Sci. Technol. 67, 102904 (2021).
    • 85. Shariat S, Badiee A, Jalali SA et al. P5 HER2/neu-derived peptide conjugated to liposomes containing MPL adjuvant as an effective prophylactic vaccine formulation for breast cancer. Cancer Lett. 355(1), 54–60 (2014). • Liposomes for prophylactic vaccine formulation for breast cancer.
    • 86. Jalali SA, Sankian M, Tavakkol-Afshari J, Jaafari MR. Induction of tumor-specific immunity by multi-epitope rat HER2/neu-derived peptides encapsulated in LPD Nanoparticles. Nanomed.: Nanotechnol. Biol. Med. 8(5), 692–701 (2012).
    • 87. Farzad N, Barati N, Momtazi-Borojeni AA et al. P435 HER2/neu-derived peptide conjugated to liposomes containing DOPE as an effective prophylactic vaccine formulation for breast cancer. Artif. Cells Nanomed. Biotechnol. 47(1), 665–673 (2019).
    • 88. Naghibi L, Yazdani M, Momtazi-Borojeni AA et al. Preparation of nanoliposomes containing HER2/neu (P5+435) peptide and evaluation of their immune responses and anti-tumoral effects as a prophylactic vaccine against breast cancer. PLOS ONE 15(12), e0243550 (2020).
    • 89. Mansourian M, Badiee A, Jalali SA et al. Effective induction of anti-tumor immunity using p5 HER-2/neu derived peptide encapsulated in fusogenic DOTAP cationic liposomes co-administrated with CpG-ODN. Immunol. Lett. 162(1 Pt A), 87–93 (2014).
    • 90. Razazan A, Behravan J, Arab A et al. Conjugated nanoliposome with the HER2/neu-derived peptide GP2 as an effective vaccine against breast cancer in mice xenograft model. PLOS ONE 12(10), e0185099 (2017).
    • 91. Vacchelli E, Galluzzi L, Eggermont A et al. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 1(6), 894–907 (2012).
    • 92. Alexander J, Sidney J, Southwood S et al. Development of high potency universal DR-restricted helper epitopes by modification of high affinity DR-blocking peptides. Immunity 1(9), 751–761 (1994).
    • 93. Zamani P, Navashenaq JG, Nikpoor AR et al. MPL nano-liposomal vaccine containing P5 HER2/neu-derived peptide pulsed PADRE as an effective vaccine in a mice TUBO model of breast cancer. J. Control Rel. 303, 223–236 (2019).
    • 94. Shen L, Li J, Liu Q et al. Local blockade of interleukin 10 and C-X-C motif chemokine ligand 12 with nano-delivery promotes antitumor response in murine cancers. ACS Nano 12(10), 9830–9841 (2018).
    • 95. Beltrán-Gracia E, López-Camacho A, Higuera-Ciapara I et al. Nanomedicine review: clinical developments in liposomal applications. Cancer Nanotechnol. 10(1), 11 (2019).
    • 96. Shi Y. Clinical translation of nanomedicine and biomaterials for cancer immunotherapy: progress and perspectives. Adv. Ther. 3(9), 1900215 (2020).
    • 97. Soliman HH. nab-Paclitaxel as a potential partner with checkpoint inhibitors in solid tumors. Oncol. Targets Ther. 10, 101–112 (2017).
    • 98. Schmid P, Adams S, Rugo HS et al. Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer. N. Engl. J. Med. 379(22), 2108–2121 (2018).
    • 99. Shroff RT, Kennedy EB, Bachini M et al. Adjuvant therapy for resected biliary tract cancer: ASCO Clinical Practice Guideline. J. Clin. Oncol. 37(12), 1015–1027 (2019).
    • 100. Badwaik HR, Kumari L, Nakhate K, Verma VS, Sakure K. Chapter 13: Phytoconstituent plumbagin: chemical, biotechnological and pharmaceutical aspects. In: Studies in Natural Products Chemistry. Atta Ur R (Ed.). Elsevier, Amsterdam, Netherlands, 415–460 (2019).
    • 101. Wang S, Hu X, Wei W, Ma G. Transformable vesicles for cancer immunotherapy. Adv. Drug Deliv. Rev. 179, 113905 (2021).
    • 102. Uchegbu IF, Vyas SP. Non-ionic surfactant based vesicles (niosomes) in drug delivery. Int. J. Pharm. 172(1), 33–70 (1998).
    • 103. Brewer JM, Alexander J. The adjuvant activity of non-ionic surfactant vesicles (niosomes) on the BALB/c humoral response to bovine serum albumin. Immunology 75(4), 570–575 (1992).
    • 104. Brewer JM, Alexander J. Studies on the adjuvant activity of non-ionic surfactant vesicles: adjuvant-driven IgG2a production independent of MHC control. Vaccine 12(7), 613–619 (1994).
    • 105. Jain S, Singh P, Mishra V, Vyas SP. Mannosylated niosomes as adjuvant-carrier system for oral genetic immunization against hepatitis B. Immunol. Lett. 101(1), 41–49 (2005).
    • 106. Jain S, Vyas SP. Mannosylated niosomes as adjuvant-carrier system for oral mucosal immunization. J. Liposome Res. 16(4), 331–345 (2006).
    • 107. Vyas SP, Singh RP, Jain S et al. Non-ionic surfactant based vesicles (niosomes) for non-invasive topical genetic immunization against hepatitis B. Int. J. Pharm. 296(1–2), 80–86 (2005).
    • 108. Jain S, Sharma RK, Vyas SP. Chitosan nanoparticles encapsulated vesicular systems for oral immunization: preparation, in-vitro and in-vivo characterization. J. Pharm. Pharmacol. 58(3), 303–310 (2006).
    • 109. Pamornpathomkul B, Niyomtham N, Yingyongnarongkul BE et al. Cationic niosomes for enhanced skin immunization of plasmid DNA-encoding ovalbumin via hollow microneedles. AAPS PharmSciTech 19(1), 481–488 (2018).
    • 110. Rathee J, Kanwar R, Kaushik D et al. Niosomes as efficient drug delivery modules for encapsulation of Toll-like receptor 7 agonists and IDO-inhibitor. Appl. Surf. Sci. 505, 144078 (2020).
    • 111. Müller RH, Mäder K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery – a review of the state of the art. Eur. J. Pharm. Biopharm. 50(1), 161–177 (2000).
    • 112. Tekade RK, Maheshwari R, Tekade M, Chougule MB. Chapter 8: solid lipid nanoparticles for targeting and delivery of drugs and genes. In: Nanotechnology-Based Approaches for Targeting and Delivery of Drugs and Genes. Mishra VKesharwani PMohd Amin MCIIyer A (Eds). Academic Press, MA, USA, 256–286 (2017).
    • 113. Talluri SV, Kuppusamy G, Karri VV et al. Lipid-based nanocarriers for breast cancer treatment – comprehensive review. Drug Deliv. 23(4), 1291–1305 (2016).
    • 114. Rajpoot K. Solid lipid nanoparticles: a promising nanomaterial in drug delivery. Curr. Pharm. Des. 25(37), 3943–3959 (2019).
    • 115. Almeida AJ, Souto E. Solid lipid nanoparticles as a drug delivery system for peptides and proteins. Adv. Drug Deliv. Rev. 59(6), 478–490 (2007).
    • 116. Stelzner JJ, Behrens M, Behrens SE, Mäder K. Squalene containing solid lipid nanoparticles, a promising adjuvant system for yeast vaccines. Vaccine 36(17), 2314–2320 (2018).
    • 117. Limeres MJ, Suñé-Pou M, Prieto-Sánchez S et al. Development and characterization of an improved formulation of cholesteryl oleate-loaded cationic solid-lipid nanoparticles as an efficient non-viral gene delivery system. Colloids Surf B Biointerfaces 184, 110533 (2019).
    • 118. Mishra H, Mishra D, Mishra PK et al. Evaluation of solid lipid nanoparticles as carriers for delivery of hepatitis B surface antigen for vaccination using subcutaneous route. J. Pharm. Pharm. Sci. 13(4), 495–509 (2010).
    • 119. Saljoughian N, Zahedifard F, Doroud D et al. Cationic solid-lipid nanoparticles are as efficient as electroporation in DNA vaccination against visceral leishmaniasis in mice. Parasite Immunol 35(12), 397–408 (2013).
    • 120. Liu Y, Tiruthani K, Wang M et al. Tumor-targeted gene therapy with lipid nanoparticles inhibits tumor-associated adipocytes and remodels the immunosuppressive tumor microenvironment in triple-negative breast cancer. Nanoscale Horiz 6(4), 319–329 (2021).
    • 121. Deng C, Jia M, Wei G et al. Inducing optimal antitumor immune response through coadministering iRGD with pirarubicin loaded nanostructured lipid carriers for breast cancer therapy. Mol. Pharm. 14(1), 296–309 (2017).
    • 122. Ganta S, Talekar M, Singh A et al. Nanoemulsions in translational research-opportunities and challenges in targeted cancer therapy. AAPS PharmSciTech 15(3), 694–708 (2014).
    • 123. Verma P, Meher JG, Asthana S et al. Perspectives of nanoemulsion assisted oral delivery of docetaxel for improved chemotherapy of cancer. Drug Deliv. 23(2), 479–488 (2016).
    • 124. Maeda H, Wu J, Sawa T et al. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J. Control Rel. 65(1–2), 271–284 (2000).
    • 125. Praveen Kumar G, Divya A. Nanoemulsion based targeting in cancer therapeutics. Med. Chem. 5(5), 272–284 (2015).
    • 126. Liu C, Lai H, Chen T. Boosting natural killer cell-based cancer immunotherapy with selenocystine/transforming growth factor-beta inhibitor-encapsulated nanoemulsion. ACS Nano 14(9), 11067–11082 (2020).
    • 127. Torchilin VP. Micellar nanocarriers: pharmaceutical perspectives. Pharm. Res. 24(1), 1–16 (2007).
    • 128. Trubetskoy VS. Polymeric micelles as carriers of diagnostic agents. Adv. Drug Deliv. Rev. 37(1–3), 81–88 (1999).
    • 129. Wei X, Liu L, Li X et al. Selectively targeting tumor-associated macrophages and tumor cells with polymeric micelles for enhanced cancer chemo-immunotherapy. J. Control Rel. 313, 42–53 (2019).
    • 130. Wang Y, Luan Z, Zhao C et al. Target delivery selective CSF-1R inhibitor to tumor-associated macrophages via erythrocyte-cancer cell hybrid membrane camouflaged pH-responsive copolymer micelle for cancer immunotherapy. Eur. J. Pharm. Sci. 142, 105136 (2020).
    • 131. Zhou L, Hou B, Wang D et al. Engineering polymeric prodrug nanoplatform for vaccination immunotherapy of cancer. Nano Lett. 20(6), 4393–4402 (2020).
    • 132. Nair PR. Delivering combination chemotherapies and targeting oncogenic pathways via polymeric drug delivery systems. Polymers 11(4), 630 (2019).
    • 133. Wong KH, Lu A, Chen X, Yang Z. Natural ingredient-based polymeric nanoparticles for cancer treatment. Molecules 25(16), 3620 (2020).
    • 134. Cafaggi S, Russo E, Stefani R et al. Preparation and evaluation of nanoparticles made of chitosan or N-trimethyl chitosan and a cisplatin-alginate complex. J. Control Rel 121(1–2), 110–123 (2007).
    • 135. Castro F, Pinto ML, Pereira CL et al. Chitosan/γ-PGA nanoparticles-based immunotherapy as adjuvant to radiotherapy in breast cancer. Biomaterials 257, 120218 (2020).
    • 136. Kim KS, Han JH, Choi SH et al. Cationic nanoparticle-mediated activation of natural killer cells for effective cancer immunotherapy. ACS Appl. Mater. Interfaces 12(51), 56731–56740 (2020).
    • 137. Masjedi A, Ahmadi A, Ghani S et al. Silencing adenosine A2a receptor enhances dendritic cell-based cancer immunotherapy. Nanomedicine 29, 102240 (2020).
    • 138. Masjedi A, Ahmadi A, Atyabi F et al. Silencing of IL-6 and STAT3 by siRNA loaded hyaluronate-N,N,N-trimethyl chitosan nanoparticles potently reduces cancer cell progression. Int. J. Biol. Macromol 149, 487–500 (2020).
    • 139. Dey A, Manna S, Kumar S et al. Immunostimulatory effect of chitosan conjugated green copper oxide nanoparticles in tumor immunotherapy. Cytokine 127, 154958 (2020).
    • 140. Bastaki S, Aravindhan S, Ahmadpour Saheb N et al. Codelivery of STAT3 and PD-L1 siRNA by hyaluronate-TAT trimethyl/thiolated chitosan nanoparticles suppresses cancer progression in tumor-bearing mice. Life Sci. 266, 118847 (2021).
    • 141. Liu L, Wang Y, Guo X et al. A biomimetic polymer magnetic nanocarrier polarizing tumor-associated macrophages for potentiating immunotherapy. Small 16(38), e2003543 (2020).
    • 142. Kakwere H, Zhang H, Ingham ES et al. Systemic immunotherapy with micellar resiquimod-polymer conjugates triggers a robust antitumor response in a breast cancer model. Adv. Healthc. Mater. 10(10), e2100008 (2021).
    • 143. Chen C, Guo Q, Fu H et al. Asynchronous blockade of PD-L1 and CD155 by polymeric nanoparticles inhibits triple-negative breast cancer progression and metastasis. Biomaterials 275, 120988 (2021).
    • 144. Kresge ACT, Leonowicz ME, Roth WJ et al. Ordered mesoporous molecular sieves synthesized by a liquid-crystal template mechanism. Nature 359(6397), 710–712 (1992).
    • 145. Chen YP, Xu L, Tang TW et al. STING Activator c-di-GMP-loaded mesoporous silica nanoparticles enhance immunotherapy against breast cancer. ACS Appl. Mater. Interfaces 12(51), 56741–56752 (2020).
    • 146. Becicka WM, Bielecki PA, Lorkowski ME et al. The effect of PEGylation on the efficacy and uptake of an immunostimulatory nanoparticle in the tumor immune microenvironment. Nanoscale Adv. 3(17), 4961–4972 (2021).
    • 147. Xu JL, Jin B, Ren ZH et al. Chemotherapy plus erlotinib versus chemotherapy alone for treating advanced non-small cell lung cancer: a meta-analysis. PLOS ONE 10(7), e0131278 (2015).
    • 148. Gurunathan S, Kang MH, Qasim M, Kim JH. Nanoparticle-mediated combination therapy: two-in-one approach for cancer. Int. J. Mol. Sci. 19(10), 3264 (2018).
    • 149. Liu R, An Y, Jia W et al. Macrophage-mimic shape changeable nanomedicine retained in tumor for multimodal therapy of breast cancer. J. Control. Rel. 321, 589–601 (2020).
    • 150. Esteva FJ, Hubbard-Lucey VM, Tang J, Pusztai L. Immunotherapy and targeted therapy combinations in metastatic breast cancer. Lancet Oncol. 20(3), e175–e186 (2019).
    • 151. Lang T, Liu Y, Zheng Z et al. Cocktail strategy based on spatio-temporally controlled nano device improves therapy of breast cancer. Adv. Mater. 31(5), e1806202 (2019).
    • 152. Zhang W, Zhang CC, Wang XY et al. Light-responsive core-shell nanoplatform for bimodal imaging-guided photothermal therapy-primed cancer immunotherapy. ACS Appl. Mater. Interfaces 12(43), 48420–48431 (2020).
    • 153. Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nat. Rev. Drug Discov. 7(9), 771–782 (2008).
    • 154. Restifo NP, Smyth MJ, Snyder A. Acquired resistance to immunotherapy and future challenges. Nat. Rev. Cancer 16(2), 121–126 (2016).
    • 155. Chemocare. Atezolizumab (Tecentriq). https://chemocare.com/chemotherapy/drug-info/atezolizumab.aspx
    • 156. Immunotherapy for Breast Cancer. How is immunotherapy for breast cancer changing the outlook for patients? Cancer Research Institute. www.cancerresearch.org/en-us/immunotherapy/cancer-types/breast-cancer
    • 157. Targeted Antibodies: mAbs, ADCs, BiTEs, and More. Monoclonal antibodies, antibody-drug conjugates, and bispecific antibodies. Cancer Resaerch Institute www.cancerresearch.org/en-us/immunotherapy/treatment-types/targeted-antibodies
    • 158. Barar J, Omidi Y. Personalized cell-mediated immunotherapy and vaccination: combating detrimental uprisings of malignancies. Bioimpacts 5(2), 65–69 (2015).
    • 159. Fathi M, Abdolahinia ED, Barar J, Omidi Y. Smart stimuli-responsive biopolymeric nanomedicines for targeted therapy of solid tumors. Nanomedicine 15(22), 2171–2200 (2020).
    • 160. Dolatkhah M, Hashemzadeh N, Barar J et al. Graphene-based multifunctional nanosystems for simultaneous detection and treatment of breast cancer. Colloids Surf. B Biointerfaces 193, 111104 (2020).
    • 161. Liu L, Wang Y, Miao L et al. Combination Immunotherapy of MUC1 mRNA Nano-vaccine and CTLA-4 Blockade Effectively Inhibits Growth of Triple Negative Breast Cancer. Mol. Ther. 26(1), 45–55 (2018).
    • 162. Lei J, Wang H, Zhu D et al. Combined effects of avasimibe immunotherapy, doxorubicin chemotherapy, and metal-organic frameworks nanoparticles on breast cancer. J. Cell Physiol. 235(5), 4814–4823 (2020). • Combination therapy.
    • 163. Yuan SJ, Xu YH, Wang C et al. Doxorubicin-polyglycerol-nanodiamond conjugate is a cytostatic agent that evades chemoresistance and reverses cancer-induced immunosuppression in triple-negative breast cancer. J. Nanobiotechnology 17(1), 110 (2019).
    • 164. Wei J, Long Y, Guo R et al. Multifunctional polymeric micelle-based chemo-immunotherapy with immune checkpoint blockade for efficient treatment of orthotopic and metastatic breast cancer. Acta Pharm. Sinica B 9(4), 819–831 (2019).
    • 165. Liu X, Feng Z, Wang C et al. Co-localized delivery of nanomedicine and nanovaccine augments the postoperative cancer immunotherapy by amplifying T-cell responses. Biomaterials 230, 119649 (2020).
    • 166. Liu Y, Qiao L, Zhang S et al. Dual pH-responsive multifunctional nanoparticles for targeted treatment of breast cancer by combining immunotherapy and chemotherapy. Acta Biomater. 66, 310–324 (2018).
    • 167. Wan Z, Sun J, Xu J et al. Dual functional immunostimulatory polymeric prodrug carrier with pendent indoximod for enhanced cancer immunochemotherapy. Acta Biomater. 90, 300–313 (2019).
    • 168. Sun JJ, Chen YC, Huang YX et al. Programmable co-delivery of the immune checkpoint inhibitor NLG919 and chemotherapeutic doxorubicin via a redox-responsive immunostimulatory polymeric prodrug carrier. Acta Pharmacol. Sinica 38(6), 823–834 (2017).
    • 169. Qin T, Xu X, Zhang Z et al. Paclitaxel/sunitinib-loaded micelles promote an antitumor response in vitro through synergistic immunogenic cell death for triple-negative breast cancer. Nanotechnology 31(36), 365101 (2020).
    • 170. Zhang LX, Sun XM, Xu ZP, Liu RT. Development of multifunctional clay-based nanomedicine for elimination of primary invasive breast cancer and prevention of its lung metastasis and distant inoculation. ACS Appl. Mater. Interfaces 11(39), 35566–35576 (2019).
    • 171. Liu J, Ai X, Cabral H, Liu J, Huang Y, Mi P. Tumor hypoxia-activated combinatorial nanomedicine triggers systemic antitumor immunity to effectively eradicate advanced breast cancer. Biomaterials 273, 120847 (2021).
    • 172. Kumar P, Srivastava R. IR 820 dye encapsulated in polycaprolactone glycol chitosan: Poloxamer blend nanoparticles for photo immunotherapy for breast cancer. Mater. Sci. Eng. C Mater. Biol. Appl. 57, 321–327 (2015).
    • 173. Cheng Y, Chen Q, Guo Z et al. An intelligent biomimetic nanoplatform for holistic treatment of metastatic triple-negative breast cancer via photothermal ablation and immune remodeling. ACS Nano 14(11), 15161–15181 (2020).
    • 174. Zhang J, Zhang D, Li Q et al. Task-specific design of immune-augmented nanoplatform to enable high-efficiency tumor immunotherapy. ACS Appl. Mater. Interfaces 11(46), 42904–42916 (2019).
    • 175. Lan Y, Liang Q, Sun Y et al. Codelivered chemotherapeutic doxorubicin via a dual-functional immunostimulatory polymeric prodrug for breast cancer immunochemotherapy. ACS Appl. Mater. Interfaces 12(28), 31904–31921 (2020).
    • 176. Luo L, Zhu C, Yin H et al. Laser immunotherapy in combination with perdurable pd-1 blocking for the treatment of metastatic tumors. ACS Nano 12(8), 7647–7662 (2018).
    • 177. 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).
    • 178. Wang L, Wang M, Zhou B et al. PEGylated reduced-graphene oxide hybridized with Fe(3)O(4) nanoparticles for cancer photothermal-immunotherapy. J. Mater. Chem. B 7(46), 7406–7414 (2019).
    • 179. Wang R, He Z, Cai P et al. Surface-functionalized modified copper sulfide nanoparticles enhance checkpoint blockade tumor immunotherapy by photothermal therapy and antigen Capturing. ACS Appl. Mater. Interfaces 11(15), 13964–13972 (2019).
    • 180. Liu H, Hu Y, Sun Y et al. Co-delivery of bee venom melittin and a photosensitizer with an organic-inorganic hybrid nanocarrier for photodynamic therapy and immunotherapy. ACS Nano 13(11), 12638–12652 (2019).
    • 181. Yang G, Xu L, Xu J et al. Smart nanoreactors for ph-responsive tumor homing, mitochondria-targeting, and enhanced photodynamic-immunotherapy of cancer. Nano Lett. 18(4), 2475–2484 (2018).
    • 182. Yu X, Gao D, Gao L et al. Inhibiting metastasis and preventing tumor relapse by triggering host immunity with tumor-targeted photodynamic therapy using photosensitizer-loaded functional nanographenes. ACS Nano 11(10), 10147–10158 (2017).
    • 183. Duan X, Chan C, Guo N, Han W, Weichselbaum RR, Lin W. Photodynamic therapy mediated by nontoxic core-shell nanoparticles synergizes with immune checkpoint blockade to elicit antitumor immunity and antimetastatic effect on breast cancer. J. Am. Chem. Soc. 138(51), 16686–16695 (2016).
    • 184. Marrache S, Tundup S, Harn DA, Dhar S. Ex vivo programming of dendritic cells by mitochondria-targeted nanoparticles to produce interferon-gamma for cancer immunotherapy. ACS Nano 7(8), 7392–7402 (2013).
    • 185. Wang M, Song J, Zhou F et al. NIR-triggered phototherapy and immunotherapy via an antigen-capturing nanoplatform for metastatic cancer treatment. Adv Sci 6(10), 1802157 (2019).
    • 186. Fatima SW, Khare SK. Benefits and challenges of antibody drug conjugates as novel form of chemotherapy. J. Control. Rel. 341, 555–565 (2022).
    • 187. Howard D, Garcia-Parra J, Healey GD et al. Antibody-drug conjugates and other nanomedicines: the frontier of gynaecological cancer treatment. Interface Focus 6(6), 20160054 (2016).
    • 188. Iyer U, Kadambi VJ. Antibody drug conjugates – Trojan horses in the war on cancer. J. Pharmacol. Toxicol. Methods 64(3), 207–212 (2011).
    • 189. De Goeij BE, Lambert JM. New developments for antibody-drug conjugate-based therapeutic approaches. Curr. Opin. Immunol. 40, 14–23 (2016).
    • 190. Takahashi I, Takahashi K, Ichimura M et al. Duocarmycin A, a new antitumor antibiotic from Streptomyces. J. Antibiot. (Tokyo) 41(12), 1915–1917 (1988).
    • 191. Matsumura Y, Kataoka K. Preclinical and clinical studies of anticancer agent-incorporating polymer micelles. Cancer Sci. 100(4), 572–579 (2009).
    • 192. Lu Y, Park K. Polymeric micelles and alternative nanonized delivery vehicles for poorly soluble drugs. Int. J. Pharm. 453(1), 198–214 (2013).