Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
ACS Appl Mater Interfaces ; 16(22): 28184-28192, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38770711

RESUMEN

B cells, despite their several unique functionalities, remain largely untapped for use as an adoptive cell therapy and are limited to in vitro use for antibody production. B cells can be easily sourced, they possess excellent lymphoid-homing capabilities, and they can act as antigen-presenting cells (APCs), offering an alternative to dendritic cells (DCs), which have shown limited efficacy in the clinical setting. Soluble factors such as IL-4 and anti-CD40 antibody can enhance the activation, survival, and antigen-presenting capabilities of B cells; however, it is difficult to attain sufficiently high concentrations of these biologics to stimulate B cells in vivo. Micropatches as Cell Engagers (MACE) are polymeric microparticles, surface functionalized with anti-CD40 and anti-IgM, which can attach to B cells and simultaneously engage multiple B-cell receptors (BCR) and CD40 receptors. Stimulation of these receptors through MACE, unlike free antibodies, enhanced the display of costimulatory molecules on the B-cell surface, increased B-cell viability, and improved antigen presentation by B cells to T cells in vitro. B-cell activation by MACE further synergized with soluble IL-4 and anti-CD40. MACE also elicited T-cell chemokine secretion by B cells. Upon intravenous adoptive transfer, MACE-bound B cells homed to the spleen and lymph nodes, key sites for antigen presentation to T cells. Adoptive transfer of MACE-B cells pulsed with the CD4+ and CD8+ epitopes of ovalbumin significantly delayed tumor progression in a murine subcutaneous EG7-OVA tumor model, demonstrating the functional benefit conferred to B cells by MACE.


Asunto(s)
Linfocitos B , Antígenos CD40 , Polímeros , Animales , Linfocitos B/inmunología , Ratones , Antígenos CD40/metabolismo , Antígenos CD40/inmunología , Polímeros/química , Receptores de Antígenos de Linfocitos B/metabolismo , Humanos , Linfocitos T/inmunología , Interleucina-4 , Ratones Endogámicos C57BL
2.
ACS Appl Mater Interfaces ; 16(22): 28070-28079, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38779939

RESUMEN

Cellular hitchhiking is an emerging strategy for the in vivo control of adoptively transferred immune cells. Hitchhiking approaches are primarily mediated by adhesion of nano and microparticles to the cell membrane, which conveys an ability to modulate transferred cells via local drug delivery. Although T cell therapies employing this strategy have progressed into the clinic, phagocytic cells including dendritic cells (DCs) are much more challenging to engineer. DC vaccines hold great potential for a spectrum of diseases, and the combination drug delivery is an attractive strategy to manipulate their function and overcome in vivo plasticity. However, DCs are not compatible with current hitchhiking approaches due to their broad phagocytic capacity. In this work, we developed and validated META (membrane engineering using tannic acid) to enable DC cellular hitchhiking for the first time. META employs the polyphenol tannic acid (TA) to facilitate supramolecular assembly of protein drug cargoes on the cell membrane, enabling the creation of cell surface-bound formulations for local drug delivery to carrier DCs. We optimized META formulations to incorporate and release protein cargoes with varying physical properties alone and in combination and to preserve DC viability and critical functions such as migration. We further show that META loaded with either a pro- or anti-inflammatory cargo can influence the carrier cell phenotype, thus demonstrating the flexibility of the approach for applications from cancer to autoimmune disease. Overall, this approach illustrates a new platform for the local control of phagocytic immune cells as a next step to advance DC therapies in the clinic.


Asunto(s)
Células Dendríticas , Polifenoles , Taninos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Taninos/química , Taninos/farmacología , Polifenoles/química , Polifenoles/farmacología , Humanos , Animales , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Ratones , Membrana Celular/metabolismo , Supervivencia Celular/efectos de los fármacos
3.
ACS Nano ; 18(15): 10439-10453, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38567994

RESUMEN

The cGAS-STING pathway plays a crucial role in innate immune activation against cancer and infections, and STING agonists based on cyclic dinucleotides (CDN) have garnered attention for their potential use in cancer immunotherapy and vaccines. However, the limited drug-like properties of CDN necessitate an efficient delivery system to the immune system. To address these challenges, we developed an immunostimulatory delivery system for STING agonists. Here, we have examined aqueous coordination interactions between CDN and metal ions and report that CDN mixed with Zn2+ and Mn2+ formed distinctive crystal structures. Further pharmaceutical engineering led to the development of a functional coordination nanoparticle, termed the Zinc-Mn-CDN Particle (ZMCP), produced by a simple aqueous one-pot synthesis. Local or systemic administration of ZMCP exerted robust antitumor efficacy in mice. Importantly, recombinant protein antigens from SARS-CoV-2 can be simply loaded during the aqueous one-pot synthesis. The resulting ZMCP antigens elicited strong cellular and humoral immune responses that neutralized SARS-CoV-2, highlighting ZMCP as a self-adjuvant vaccine platform against COVID-19 and other infectious pathogens. Overall, this work establishes a paradigm for developing translational coordination nanomedicine based on drug-metal ion coordination and broadens the applicability of coordination medicine for the delivery of proteins and other biologics.


Asunto(s)
Nanopartículas , Neoplasias , Vacunas , Animales , Ratones , Neoplasias/terapia , Adyuvantes Inmunológicos , Inmunoterapia/métodos , Nanopartículas/química
4.
Adv Healthc Mater ; : e2304144, 2024 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-38581301

RESUMEN

Adoptive cell therapies are dramatically altering the treatment landscape of cancer. However, treatment of solid tumors remains a major unmet need, in part due to limited adoptive cell infiltration into the tumor and in part due to the immunosuppressive tumor microenvironment. The heterogeneity of tumors and presence of nonresponders also call for development of antigen-independent therapeutic approaches. Myeloid cells offer such an opportunity, given their large presence in the immunosuppressive tumor microenvironment, such as in triple negative breast cancer. However, their therapeutic utility is hindered by their phenotypic plasticity. Here, the impressive trafficking ability of adoptively transferred monocytes is leveraged into the immunosuppressive 4T1 tumor to develop an antitumor therapy. To control monocyte differentiation in the tumor microenvironment, surface-adherent "backpacks" stably modified with interferon gamma (IFNγ) are developed to stimulate macrophage plasticity into a pro-inflammatory, antitumor phenotype, a strategy as referred to as Ornate Polymer backpacks on Tissue Infiltrating Monocytes (OPTIMs). Treatment with OPTIMs substantially reduces tumor burden in a mouse 4T1 model and significantly increases survival. Cytokine and immune cell profiling reveal that OPTIMs remodeled the tumor microenvironment into a pro-inflammatory state.

5.
Nat Biomed Eng ; 8(5): 579-592, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38424352

RESUMEN

Tumour-associated neutrophils can exert antitumour effects but can also assume a pro-tumoural phenotype in the immunosuppressive tumour microenvironment. Here we show that neutrophils can be polarized towards the antitumour phenotype by discoidal polymer micrometric 'patches' that adhere to the neutrophils' surfaces without being internalized. Intravenously administered micropatch-loaded neutrophils accumulated in the spleen and in tumour-draining lymph nodes, and activated splenic natural killer cells and T cells, increasing the accumulation of dendritic cells and natural killer cells. In mice bearing subcutaneous B16F10 tumours or orthotopic 4T1 tumours, intravenous injection of the micropatch-loaded neutrophils led to robust systemic immune responses, a reduction in tumour burden and improvements in survival rates. Micropatch-activated neutrophils combined with the checkpoint inhibitor anti-cytotoxic T-lymphocyte-associated protein 4 resulted in strong inhibition of the growth of B16F10 tumours, and in complete tumour regression in one-third of the treated mice. Micropatch-loaded neutrophils could provide a potent, scalable and drug-free approach for neutrophil-based cancer immunotherapy.


Asunto(s)
Inmunoterapia , Ratones Endogámicos C57BL , Neutrófilos , Polímeros , Animales , Neutrófilos/inmunología , Inmunoterapia/métodos , Ratones , Polímeros/química , Línea Celular Tumoral , Microambiente Tumoral/efectos de los fármacos , Femenino , Ratones Endogámicos BALB C , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Melanoma Experimental/patología , Neoplasias/inmunología , Neoplasias/terapia , Células Asesinas Naturales/inmunología , Humanos
6.
Bioeng Transl Med ; 9(1): e10588, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38193112

RESUMEN

Vaccines are an important tool in the rapidly evolving repertoire of immunotherapies in oncology. Although cancer vaccines have been investigated for over 30 years, very few have achieved meaningful clinical success. However, recent advances in areas such antigen identification, formulation development and manufacturing, combination therapy regimens, and indication and patient selection hold promise to reinvigorate the field. Here, we provide a timely update on the clinical status of cancer vaccines. We identify and critically analyze 360 active trials of cancer vaccines according to delivery vehicle, antigen type, indication, and other metrics, as well as highlight eight globally approved products. Finally, we discuss current limitations and future applications for clinical translation of cancer vaccines.

7.
Sci Transl Med ; 16(728): eadk5413, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38170792

RESUMEN

The choroid plexus (ChP) of the brain plays a central role in orchestrating the recruitment of peripheral leukocytes into the central nervous system (CNS) through the blood-cerebrospinal fluid (BCSF) barrier in pathological conditions, thus offering a unique niche to diagnose CNS disorders. We explored whether magnetic resonance imaging of the ChP could be optimized for mild traumatic brain injury (mTBI). mTBI induces subtle, yet influential, changes in the brain and is currently severely underdiagnosed. We hypothesized that mTBI induces sufficient alterations in the ChP to cause infiltration of circulating leukocytes through the BCSF barrier and developed macrophage-adhering gadolinium [Gd(III)]-loaded anisotropic micropatches (GLAMs), specifically designed to image infiltrating immune cells. GLAMs are hydrogel-based discoidal microparticles that adhere to macrophages without phagocytosis. We present a fabrication process to prepare GLAMs at scale and demonstrate their loading with Gd(III) at high relaxivities, a key indicator of their effectiveness in enhancing image contrast and clarity in medical imaging. In vitro experiments with primary murine and porcine macrophages demonstrated that GLAMs adhere to macrophages also under shear stress and did not affect macrophage viability or functions. Studies in a porcine mTBI model confirmed that intravenously administered macrophage-adhering GLAMs provide a differential signal in the ChP and lateral ventricles at Gd(III) doses 500- to 1000-fold lower than those used in the current clinical standard Gadavist. Under the same mTBI conditions, Gadavist did not offer a differential signal at clinically used doses. Our results suggest that macrophage-adhering GLAMs could facilitate mTBI diagnosis.


Asunto(s)
Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , Animales , Ratones , Porcinos , Gadolinio , Lesiones Traumáticas del Encéfalo/diagnóstico por imagen , Encéfalo/patología , Imagen por Resonancia Magnética/métodos , Conmoción Encefálica/patología , Macrófagos/patología
8.
ACS Nano ; 17(16): 15918-15930, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37565806

RESUMEN

Natural killer (NK) cell therapies have emerged as a potential therapeutic approach to various cancers. Their efficacy, however, is limited by their low persistence and anergy. Current approaches to sustain NK cell persistence in vivo include genetic modification, activation via pretreatment, or coadministration of supporting cytokines or antibodies. Such supporting therapies exhibit limited efficacy in vivo, in part due to the reversal of their effect within the immunosuppressive tumor microenvironment and off-target toxicity. Here, we report a material-based approach to address this challenge. Specifically, we describe the use of polymeric micropatches as a platform for sustained, targeted activation of NK cells, an approach referred to as microparticles as cell engagers (MACE). Poly(lactide-co-glycolic) acid (PLGA) micropatches, 4-8 µm in diameter and surface-modified with NK cell receptor targeting antibodies, exhibited strong adhesion to NK cells and induced their activation without the need of coadministered cytokines. The activation induced by MACE was greater than that induced by nanoparticles, attesting to the crucial role of MACE geometry in the activation of NK cells. MACE-bound NK cells remained viable and exhibited trans-endothelial migration and antitumor activity in vitro. MACE-bound NK cells activated T cells, macrophages, and dendritic cells in vitro. Adoptive transfer of NK-MACE also demonstrated superior antitumor efficacy in a mouse melanoma lung metastasis model compared to unmodified NK cells. Overall, MACE offers a simple, scalable, and effective way of activating NK cells and represents an attractive platform to improve the efficacy of NK cell therapy.


Asunto(s)
Melanoma , Neoplasias , Animales , Ratones , Polímeros/metabolismo , Células Asesinas Naturales , Neoplasias/metabolismo , Inmunoterapia Adoptiva , Melanoma/metabolismo , Citocinas/metabolismo , Microambiente Tumoral
9.
J Control Release ; 357: 84-93, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36948420

RESUMEN

Cyclic dinucleotides (CDNs), as one type of Stimulator of Interferon Genes (STING) pathway agonist, have shown promising results for eliciting immune responses against cancer and viral infection. However, the suboptimal drug-like properties of conventional CDNs, including their short in vivo half-life and poor cellular permeability, compromise their therapeutic efficacy. In this study, we have developed a manganese-silica nanoplatform (MnOx@HMSN) that enhances the adjuvant effects of CDN by achieving synergy with Mn2+ for vaccination against cancer and SARS-CoV-2. MnOx@HMSN with large mesopores were efficiently co-loaded with CDN and peptide/protein antigens. MnOx@HMSN(CDA) amplified the activation of the STING pathway and enhanced the production of type-I interferons and other proinflammatory cytokines from dendritic cells. MnOx@HMSN(CDA) carrying cancer neoantigens elicited robust antitumor T-cell immunity with therapeutic efficacy in two different murine tumor models. Furthermore, MnOx@HMSN(CDA) loaded with SARS-CoV-2 antigen achieved strong and durable (up to one year) humoral immune responses with neutralizing capability. These results demonstrate that MnOx@HMSN(CDA) is a versatile nanoplatform for vaccine applications.


Asunto(s)
COVID-19 , Neuropatía Hereditaria Motora y Sensorial , Nanopartículas , Vacunas , Humanos , Animales , Ratones , Manganeso , Dióxido de Silicio , COVID-19/prevención & control , SARS-CoV-2 , Inmunoterapia
10.
Nat Biomed Eng ; 7(1): 72-84, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36564626

RESUMEN

The effectivity of cancer immunotherapies is hindered by immunosuppressive tumour microenvironments that are poorly infiltrated by effector T cells and natural killer cells. In infection and autoimmune disease, the recruitment and activation of effector immune cells is coordinated by pro-inflammatory T helper 17 (TH17) cells. Here we show that pathogen-mimicking hollow nanoparticles displaying mannan (a polysaccharide that activates TH17 cells in microbial cell walls) limit the fraction of regulatory T cells and induce TH17-cell-mediated anti-tumour responses. The nanoparticles activate the pattern-recognition receptor Dectin-2 and Toll-like receptor 4 in dendritic cells, and promote the differentiation of CD4+ T cells into the TH17 phenotype. In mice, intra-tumoural administration of the nanoparticles decreased the fraction of regulatory T cells in the tumour while markedly increasing the fractions of TH17 cells (and the levels of TH17-cell-associated cytokines), CD8+ T cells, natural killer cells and M1-like macrophages. The anti-tumoural activity of the effector cells was amplified by an agonistic antibody against the co-stimulatory receptor OX40 in multiple mouse models. Nanomaterials that induce TH17-cell-mediated immune responses may have therapeutic potential.


Asunto(s)
Linfocitos T CD8-positivos , Nanopartículas , Animales , Ratones , Diferenciación Celular , Citocinas , Linfocitos T Reguladores , Células Th17/inmunología
11.
J Control Release ; 352: 1093-1103, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36351520

RESUMEN

Various anti-tumor nanomedicines have been developed based on the enhanced permeability and retention effect. However, the dense extracellular matrix (ECM) in tumors remains a major barrier for the delivery and accumulation of nanoparticles into tumors. While ECM-degrading enzymes, such as collagenase, hyaluronidase, and bromelain, have been used to facilitate the accumulation of nanoparticles, serious side effects arising from the current non-tumor-specific delivery methods limit their clinical applications. Here, we report targeted delivery of bromelain into tumor tissues through its covalent attachment to a hyaluronic acid (HA)-peptide conjugate with tumor ECM targeting ability. The ECM targeting peptide, collagen type IV-binding peptide (C4BP), was chosen from six candidate-peptides based on their ability to bind to frozen sections of triple-negative breast cancer, 4T1 tumor ex vivo. The HA- C4BP conjugate showed a significant increase in tumor accumulation in 4T1-bearing mice after intravenous administration compared to unmodified HA. We further demonstrated that the systemic administration of bromelain conjugated C4BP-HA (C4BP-HA-Bro) potentiates the anti-tumor efficacy of liposomal doxorubicin. C4BP-HA-Bro decreased the number and length of collagen fibers and improved the distribution of doxorubicin within the tumor. No infusion reaction was noted after delivery of C4BP-HA-Bro. C4BP-HA thus offers a potential for effective and safe delivery of bromelain for improved intratumoral delivery of therapeutics.


Asunto(s)
Nanopartículas , Neoplasias , Ratones , Animales , Liposomas/uso terapéutico , Bromelaínas/uso terapéutico , Doxorrubicina/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Ácido Hialurónico/uso terapéutico , Nanopartículas/uso terapéutico , Péptidos/uso terapéutico , Matriz Extracelular , Línea Celular Tumoral
12.
Nat Nanotechnol ; 16(11): 1260-1270, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34594005

RESUMEN

Nutritional metal ions play critical roles in many important immune processes. Hence, the effective modulation of metal ions may open up new forms of immunotherapy, termed as metalloimmunotherapy. Here, we demonstrate a prototype of cancer metalloimmunotherapy using cyclic dinucleotide (CDN) stimulator of interferon genes (STING) agonists and Mn2+. We screened various metal ions and discovered specific metal ions augmented STING agonist activity, wherein Mn2+ promoted a 12- to 77-fold potentiation effect across the prevalent human STING haplotypes. Notably, Mn2+ coordinated with CDN STING agonists to self-assemble into a nanoparticle (CDN-Mn2+ particle, CMP) that effectively delivered STING agonists to immune cells. The CMP, administered either by local intratumoural or systemic intravenous injection, initiated robust anti-tumour immunity, achieving remarkable therapeutic efficacy with minute doses of STING agonists in multiple murine tumour models. Overall, the CMP offers a new platform for local and systemic cancer treatments, and this work underscores the great potential of coordination nanomedicine for metalloimmunotherapy.


Asunto(s)
Inmunoterapia , Manganeso/farmacología , Neoplasias/tratamiento farmacológico , Nucleótidos/farmacología , Animales , Haplotipos/efectos de los fármacos , Humanos , Inmunidad/efectos de los fármacos , Iones/química , Iones/inmunología , Iones/farmacología , Manganeso/química , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Metales/química , Metales/inmunología , Metales/farmacología , Ratones , Nanopartículas/química , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Nucleótidos/química
13.
Adv Ther (Weinh) ; 4(8)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34485685

RESUMEN

Photothermal therapy (PTT) and neoantigen cancer vaccine each offers minimally invasive and highly specific cancer therapy; however, they are not effective against large established tumors due to physical and biological barriers that attenuate thermal ablation and abolish anti-tumor immunity. Here, we designed and performed comparative study using small (~ 50 mm3) and large (> 100 mm3) tumors to examine how tumor size affects the therapeutic efficiency of PTT and neoantigen cancer vaccine. We show that spiky gold nanoparticle (SGNP)-based PTT and synergistic dual adjuvant-based neoantigen cancer vaccine can efficiently regress small tumors as a single agent, but not large tumors due to limited internal heating and immunosuppressive tumor microenvironment (TME). We report that PTT sensitizes tumors to neoantigen cancer vaccination by destroying and compromising the TME via thermally induced cellular and molecular damage, while neoantigen cancer vaccine reverts local immune suppression induced by PTT and shapes residual TME in favor of anti-tumor immunity. The combination therapy efficiently eradicated large local tumors and also exerted strong abscopal effect against pre-established distant tumors with robust systemic anti-tumor immunity. Thus, PTT combined with neoantigen cancer vaccine is a promising nano-immunotherapy for personalized therapy of advanced cancer.

14.
Biomaterials ; 274: 120844, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33962217

RESUMEN

Identification of tumor-specific mutations, called neoantigens, offers new exciting opportunities for personalized cancer immunotherapy. However, it remains challenging to achieve robust induction of neoantigen-specific T cells and drive their infiltration into the tumor microenvironment (TME). Here, we have developed a novel polyethyleneimine (PEI)-based personalized vaccine platform carrying neoantigen peptides and CpG adjuvants in a compact nanoparticle (NP) for their spatio-temporally concerted delivery. The NP vaccine significantly enhanced activation and antigen cross-presentation of dendritic cells, resulting in strong priming of neoantigen-specific CD8+ T cells with the frequency in the systemic circulation reaching as high as 23 ± 7% after a single subcutaneous administration. However, activated CD8+ T cells in circulation exhibited limited tumor infiltration, leading to poor anti-tumor efficacy. Notably, local administration of stimulator of interferon genes (STING) agonist promoted tumor infiltration of vaccine-primed CD8+ T cells, thereby overcoming one of the major challenges in achieving strong anti-tumor efficacy with cancer vaccination. The NP vaccination combined with STING agonist therapy eliminated tumors in murine models of MC-38 colon carcinoma and B16F10 melanoma and established long-term immunological memory. Our approach provides a novel therapeutic strategy based on combination nano-immunotherapy for personalized cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Vacunas contra el Cáncer , Animales , Antígenos de Neoplasias , Inmunoterapia , Ratones , Microambiente Tumoral
15.
Adv Sci (Weinh) ; 8(5): 2002577, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33717838

RESUMEN

Nanoparticles (NPs) can serve as a promising vaccine delivery platform for improving pharmacological property and codelivery of antigens and adjuvants. However, NP-based vaccines are generally associated with complex synthesis and postmodification procedures, which pose technical and manufacturing challenges for tailor-made vaccine production. Here, modularly programmed, polyethyleneimine (PEI)-based NP vaccines are reported for simple production of personalized cancer vaccines. Briefly, PEI is conjugated with neoantigens by facile coupling chemistry, followed by electrostatic assembly with CpG adjuvants, leading to the self-assembly of nontoxic, sub-50 nm PEI NPs. Importantly, PEI NPs promote activation and antigen cross-presentation of antigen-presenting cells and cross-priming of neoantigen-specific CD8+ T cells. Surprisingly, after only a single intratumoral injection, PEI NPs with optimal PEGylation elicit as high as ≈30% neoantigen-specific CD8+ T cell response in the systemic circulation and sustain elevated CD8+ T cell response over 3 weeks. PEI-based nanovaccines exert potent antitumor efficacy against pre-established local tumors as well as highly aggressive metastatic tumors. PEI engineering for modular incorporation of neoantigens and adjuvants offers a promising strategy for rapid and facile production of personalized cancer vaccines.

16.
ACS Nano ; 14(10): 13268-13278, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-32902245

RESUMEN

Therapeutic cancer vaccines require robust cellular immunity for the efficient killing of tumor cells, and recent advances in neoantigen discovery may provide safe and promising targets for cancer vaccines. However, elicitation of T cells with strong antitumor efficacy requires intricate multistep processes that have been difficult to attain with traditional vaccination approaches. Here, a multifunctional nanovaccine platform has been developed for direct delivery of neoantigens and adjuvants to lymph nodes (LNs) and highly efficient induction of neoantigen-specific T cell responses. A PEGylated reduced graphene oxide nanosheet (RGO-PEG, 20-30 nm in diameter) is a highly modular and biodegradable platform for facile preparation of neoantigen vaccines within 2 h. RGO-PEG exhibits rapid, efficient (15-20% ID/g), and sustained (up to 72 h) accumulation in LNs, achieving >100-fold improvement in LN-targeted delivery, compared with soluble vaccines. Moreover, RGO-PEG induces intracellular reactive oxygen species in dendritic cells, guiding antigen processing and presentation to T cells. Importantly, a single injection of RGO-PEG vaccine elicits potent neoantigen-specific T cell responses lasting up to 30 days and eradicates established MC-38 colon carcinoma. Further combination with anti-PD-1 therapy achieved great therapeutic improvements against B16F10 melanoma. RGO-PEG may serve a powerful delivery platform for personalized cancer vaccination.


Asunto(s)
Vacunas contra el Cáncer , Grafito , Neoplasias , Ganglios Linfáticos , Neoplasias/tratamiento farmacológico , Especies Reactivas de Oxígeno
17.
Adv Ther (Weinh) ; 3(10)2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34295963

RESUMEN

Stimulator of interferon genes (STING) activation by intratumoral STING agonist treatment has been recently shown to eradicate tumors in preclinical models of cancer immunotherapy, generating intense research interest and leading to multiple clinical trials. However, there are many challenges associated with STING agonist-based cancer immunotherapy, including low cellular uptake of STING agonists. Here, biodegradable mesoporous silica nanoparticles (bMSN) with an average size of 80 nm are developed for efficient cellular delivery of STING agonists. STING agonists delivered via bMSN potently activate innate and adaptive immune cells, leading to strong antitumor efficacy and prolonged animal survival in murine models of melanoma. Delivery of immunotherapeutic agents via biodegradable bMSN is a promising approach for improving cancer immunotherapy.

18.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31619584

RESUMEN

Lung cancer remains the leading cause of cancer-related death in the United States. Although the alveolar macrophage (AM) comprises the major resident immune cell in the lung, few studies have investigated its role in lung cancer development. We recently discovered a potentially novel mechanism wherein AMs regulate STAT-induced inflammatory responses in neighboring epithelial cells (ECs) via secretion and delivery of suppressors of cytokine signaling 3 (SOCS3) within extracellular vesicles (EVs). Here, we explored the impact of SOCS3 transfer on EC tumorigenesis and the integrity of AM SOCS3 secretion during development of lung cancer. AM-derived EVs containing SOCS3 inhibited STAT3 activation as well as proliferation and survival of lung adenocarcinoma cells. Levels of secreted SOCS3 were diminished in lungs of patients with non-small cell lung cancer and in a mouse model of lung cancer, and the impaired ability of murine AMs to secrete SOCS3 within EVs preceded the development of lung tumors. Loss of this homeostatic brake on tumorigenesis prompted our effort to "rescue" it. Provision of recombinant SOCS3 loaded within synthetic liposomes inhibited proliferation and survival of lung adenocarcinoma cells in vitro as well as malignant transformation of normal ECs. Intratumoral injection of SOCS3 liposomes attenuated tumor growth in a lung cancer xenograft model. This work identifies AM-derived vesicular SOCS3 as an endogenous antitumor mechanism that is disrupted within the tumor microenvironment and whose rescue by synthetic liposomes can be leveraged as a potential therapeutic strategy for lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Neoplasias Pulmonares/inmunología , Macrófagos Alveolares/inmunología , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Células A549 , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/inmunología , Carcinogénesis/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/metabolismo , Femenino , Humanos , Inyecciones Intralesiones , Liposomas , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Macrófagos Alveolares/citología , Macrófagos Alveolares/metabolismo , Ratones , Cultivo Primario de Células , Ratas , Proteínas Recombinantes/administración & dosificación , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Factor de Transcripción STAT3/inmunología , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/administración & dosificación , Proteína 3 Supresora de la Señalización de Citocinas/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Nat Biomed Eng ; 3(10): 768-782, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31406259

RESUMEN

Research into the immunological processes implicated in cancer has yielded a basis for the range of immunotherapies that are now considered the fourth pillar of cancer treatment (alongside surgery, radiotherapy and chemotherapy). For some aggressive cancers, such as advanced non-small-cell lung carcinoma, combination immunotherapies have resulted in unprecedented treatment efficacy for responding patients, and have become frontline therapies. Individualized immunotherapy, enabled by the identification of patient-specific mutations, neoantigens and biomarkers, and facilitated by advances in genomics and proteomics, promises to broaden the responder patient population. In this Perspective, we give an overview of immunotherapies leveraging engineering approaches, including the design of biomaterials, delivery strategies and nanotechnology solutions, for the realization of individualized cancer treatments such as nanoparticle vaccines customized with neoantigens, cell therapies based on patient-derived dendritic cells and T cells, and combinations of theranostic strategies. Developments in precision cancer immunotherapy will increasingly rely on the adoption of engineering principles.


Asunto(s)
Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/inmunología , Vacunas contra el Cáncer/inmunología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Inmunoterapia/métodos , Medicina de Precisión/métodos , Algoritmos , Animales , Antígenos de Neoplasias/genética , Antineoplásicos , Biomarcadores de Tumor/genética , Vacunas contra el Cáncer/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Tratamiento Basado en Trasplante de Células y Tejidos , Células Dendríticas/inmunología , Sistemas de Liberación de Medicamentos , Quimioterapia , Quimioterapia Combinada , Vectores Genéticos , Humanos , Mutación , Nanopartículas , Nanotecnología , Fototerapia , Linfocitos T/inmunología , Nanomedicina Teranóstica , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
20.
Biomaterials ; 217: 119287, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31247511

RESUMEN

Nucleic acids have both extensive physiological function and structural potential, rendering them quintessential engineering biomaterials. As carriers of precisely-tunable genetic information, both DNA and RNA can be synthetically generated to form a myriad of structures and to transmit specific genetic code. Importantly, recent studies have shown that DNA and RNA, both in their native and engineered forms, can function as potent regulators of innate immunity, capable of initiating and modulating immune responses. In this review, we highlight recent advances in biomaterials inspired by the various interactions of nucleic acids and the immune system. We discuss key advances in self-assembled structures based on exogenous nucleic acids and engineering approaches to apply endogenous nucleic acids as found in immunogenic cell death and extracellular traps. In addition, we discuss new strategies to control dinucleotide signaling and provide recent examples of biomaterials designed for cancer immunotherapy with STING agonists.


Asunto(s)
Biomimética , Inmunomodulación , Ácidos Nucleicos/química , Ácidos Nucleicos/inmunología , Animales , Humanos , Liposomas , Polímeros/química , Sistemas de Mensajero Secundario
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA