A melanin-mediated cancer immunotherapy patch

Transdermal microneedle patch integrated with whole tumor lysate containing melanin facilitates cancer immunotherapy upon near-infrared light irradiation. Lighting up antitumor responses Dendritic cell (DC)–based and whole tumor antigen vaccines have shown promise as cancer therapies but have been constrained by suboptimal antigen presentation and T cell activation. Ye et al. now demonstrate how a transdermal microneedle patch loaded with tumor lysate and melanin can lead to improved antitumor vaccine responses. They treated mice transdermally with a microneedle patch loaded with whole tumor lysate from B16F10 melanoma combined with melanin and exposed the patch to near-infrared light irradiation, which promoted heat generation by melanin and enhanced antigen uptake by DCs. This light-triggered heat response enhanced T cell migration and localized proinflammatory cytokine production and was effective at targeting both primary and distal secondary tumors. Melanin is capable of transforming 99.9% of the absorbed sunlight energy into heat, reducing the risk of skin cancer. We here develop a melanin-mediated cancer immunotherapy strategy through a transdermal microneedle patch. B16F10 whole tumor lysate containing melanin is loaded into polymeric microneedles that allow sustained release of the lysate upon insertion into the skin. In combination with the near-infrared light irradiation, melanin in the patch mediates the generation of heat, which further promotes tumor-antigen uptake by dendritic cells, and leads to enhanced antitumor vaccination. We found that the spatiotemporal photoresponsive immunotherapy increases infiltration of polarized T cells and local cytokine release. These immunological effects increase the survival of mice after tumor challenge and elicited antitumor effects toward established primary tumor and distant tumor. Collectively, melanin generates local heat, boosts T cell activities by transdermal vaccines, and promotes antitumor immune responses.

[1]  Wei Huang,et al.  Engineering Melanin Nanoparticles as an Efficient Drug–Delivery System for Imaging‐Guided Chemotherapy , 2015, Advanced materials.

[2]  D. Tyler,et al.  Melanoma-Derived Wnt 5 a Promotes Local Dendritic-Cell Expression of IDO and Immunotolerance : Opportunities for PharmacologicEnhancementof Immunotherapy , 2015 .

[3]  A. Filson,et al.  Isolation of Melanin Granules , 1957, Nature.

[4]  J. Stockman,et al.  Dissolving polymer microneedle patches for influenza vaccination , 2012 .

[5]  David J. Mooney,et al.  Biomaterials and emerging anticancer therapeutics: engineering the microenvironment , 2015, Nature Reviews Cancer.

[6]  Ligeng Xu,et al.  Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy , 2016, Nature Communications.

[7]  D. Irvine,et al.  Active targeting of chemotherapy to disseminated tumors using nanoparticle-carrying T cells , 2015, Science Translational Medicine.

[8]  A. J. Lea Solubility of Melanins , 1952, Nature.

[9]  G. Coukos,et al.  Whole Tumor Antigen Vaccines: Where Are We? , 2015, Vaccines.

[10]  David J Mooney,et al.  In Situ Regulation of DC Subsets and T Cells Mediates Tumor Regression in Mice , 2009, Science Translational Medicine.

[11]  Zhen Gu,et al.  Leveraging Physiology for Precision Drug Delivery , 2017 .

[12]  Darrell J Irvine,et al.  Engineering synthetic vaccines using cues from natural immunity. , 2013, Nature materials.

[13]  S. Rosenberg,et al.  Adoptive cell transfer: a clinical path to effective cancer immunotherapy , 2008, Nature Reviews Cancer.

[14]  J. Castle,et al.  Exploiting the mutanome for tumor vaccination. , 2012, Cancer research.

[15]  L. Pease,et al.  Co-potentiation of antigen recognition: A mechanism to boost weak T cell responses and provide immunotherapy in vivo , 2015, Science Advances.

[16]  S. Calderwood,et al.  The Role of Heat Shock Proteins in Antigen Cross Presentation , 2012, Front. Immun..

[17]  T. Kupper,et al.  Immune surveillance in the skin: mechanisms and clinical consequences , 2004, Nature Reviews Immunology.

[18]  S. B. Stephan,et al.  Biopolymer implants enhance the efficacy of adoptive T cell therapy , 2014, Nature Biotechnology.

[19]  John D Lambris,et al.  Complement: a key system for immune surveillance and homeostasis , 2010, Nature Immunology.

[20]  R. DePinho,et al.  BRafV600E cooperates with Pten silencing to elicit metastatic melanoma , 2009, Nature Genetics.

[21]  Martin F. Bachmann,et al.  Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns , 2010, Nature Reviews Immunology.

[22]  P. Choyke,et al.  Spatially selective depletion of tumor-associated regulatory T cells with near-infrared photoimmunotherapy , 2016, Science Translational Medicine.

[23]  Zhen Gu,et al.  Enhanced Cancer Immunotherapy by Microneedle Patch-Assisted Delivery of Anti-PD1 Antibody. , 2016, Nano letters.

[24]  Jennifer Couzin-Frankel,et al.  Breakthrough of the year 2013. Cancer immunotherapy. , 2013, Science.

[25]  Soong Ho Um,et al.  Therapeutic cell engineering using surface-conjugated synthetic nanoparticles , 2010, Nature Medicine.

[26]  M. Northridge,et al.  Who are we? , 2008, American journal of public health.

[27]  E. Mardis,et al.  A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells , 2015, Science.

[28]  D. Pardoll,et al.  STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade , 2015, Science Translational Medicine.

[29]  Zhen Gu,et al.  Microneedle-array patches loaded with hypoxia-sensitive vesicles provide fast glucose-responsive insulin delivery , 2015, Proceedings of the National Academy of Sciences.

[30]  Abhishek D. Garg,et al.  Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell–driven rejection of high-grade glioma , 2016, Science Translational Medicine.

[31]  Youngjin Choi,et al.  Injectable, spontaneously assembling inorganic scaffolds modulate immune cells in vivo and increase vaccine efficacy , 2014, Nature Biotechnology.

[32]  Clare L. Bennett,et al.  Murine epidermal Langerhans cells and langerin-expressing dermal dendritic cells are unrelated and exhibit distinct functions , 2009, Proceedings of the National Academy of Sciences.

[33]  Steven A. Rosenberg,et al.  Adoptive-cell-transfer therapy for the treatment of patients with cancer , 2003, Nature Reviews Cancer.

[34]  A. Palucka,et al.  Dendritic cells as therapeutic vaccines against cancer , 2005, Nature Reviews Immunology.

[35]  M. Sadelain,et al.  The ABCs of artificial antigen presentation , 2004, Nature Biotechnology.

[36]  M. Dhodapkar,et al.  Induction of Antigen-Specific Immunity with a Vaccine Targeting NY-ESO-1 to the Dendritic Cell Receptor DEC-205 , 2014, Science Translational Medicine.

[37]  Charles G. Drake,et al.  Breathing new life into immunotherapy: review of melanoma, lung and kidney cancer , 2014, Nature Reviews Clinical Oncology.

[38]  Liang Cheng,et al.  Functional nanomaterials for phototherapies of cancer. , 2014, Chemical reviews.

[39]  R. Teague,et al.  Adapting Cancer Immunotherapy Models for the Real World. , 2016, Trends in immunology.

[40]  Elizabeth A. Repasky,et al.  Fever and the thermal regulation of immunity: the immune system feels the heat , 2015, Nature Reviews Immunology.

[41]  D. Tyler,et al.  Melanoma-Derived Wnt5a Promotes Local Dendritic-Cell Expression of IDO and Immunotolerance: Opportunities for Pharmacologic Enhancement of Immunotherapy , 2015, Cancer Immunology Research.

[42]  Yuqi Zhang,et al.  Photoacoustic Drug Delivery , 2017, Sensors.

[43]  Nuria Oliva,et al.  Local triple-combination therapy results in tumour regression and prevents recurrence in a colon cancer model. , 2016, Nature materials.

[44]  D. Littman Releasing the Brakes on Cancer Immunotherapy , 2015, Cell.

[45]  Quanyin Hu,et al.  Tailoring Biomaterials for Cancer Immunotherapy: Emerging Trends and Future Outlook , 2017, Advanced materials.

[46]  A. Oxenius,et al.  Direct activation of antigen-presenting cells is required for CD8+ T-cell priming and tumor vaccination , 2011, Proceedings of the National Academy of Sciences.

[47]  Maria Raposo,et al.  Polymeric scaffolds for enhanced stability of melanin incorporated in liposomes. , 2010, Journal of colloid and interface science.

[48]  M. Smyth,et al.  Immune surveillance of tumors. , 2007, The Journal of clinical investigation.