Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Int J Pharm ; 656: 124045, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38561134

RESUMEN

The field of cancer therapy is witnessing the emergence of immunotherapy, an innovative approach that activates the body own immune system to combat cancer. Immunogenic cell death (ICD) has emerged as a prominent research focus in the field of cancer immunotherapy, attracting significant attention in recent years. The activation of ICD can induce the release of damage-associated molecular patterns (DAMPs), such as calreticulin (CRT), adenosine triphosphate (ATP), high mobility group box protein 1 (HMGB1), and heat shock proteins (HSP). Subsequently, this process promotes the maturation of innate immune cells, including dendritic cells (DCs), thereby triggering a T cell-mediated anti-tumor immune response. The activation of the ICD ultimately leads to the development of long-lasting immune responses against tumors. Studies have demonstrated that partial therapeutic approaches, such as chemotherapy with doxorubicin, specific forms of radiotherapy, and phototherapy, can induce the generation of ICD. The main focus of this article is to discuss and review the therapeutic methods triggered by nanoparticles for ICD, while briefly outlining their anti-tumor mechanism. The objective is to provide a comprehensive reference for the widespread application of ICD.


Asunto(s)
Muerte Celular Inmunogénica , Inmunoterapia , Nanopartículas , Neoplasias , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Neoplasias/terapia , Neoplasias/inmunología , Neoplasias/tratamiento farmacológico , Inmunoterapia/métodos , Animales , Nanopartículas/administración & dosificación , Células Dendríticas/inmunología , Células Dendríticas/efectos de los fármacos
2.
Phytomedicine ; 128: 155504, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38452404

RESUMEN

BACKGROUND: Pinellia pedatisecta Schott extract (PE) is extracted from Pinellia pedatisecta Schott (PPS), a traditional Chinese medicinal plant with the potential for direct anticancer effects or eliciting an anti-tumor response by activating the immune system. PURPOSE: To explore PE's ability and mechanism to reconstruct cisplatin's immunogenicity. METHODS: Cervical cancer cells were treated with cisplatin (CDDP) and/or PE. The exposure of calreticulin (CRT) on cell membrane was investigated by flow cytometry. The extracellular of ATP and HMGB1 was investigated by Western blot analysis, immunofluorescence and ELISA assay. Changes in immune profiles were using flow cytometry in vaccination and anti-tumor assays in vivo. Lastly, the mechanism of PE influenced the ROS/ERS pathway was examined by ROS assay kit, flow cytometry and Western blotting. RESULTS: PE treatment induced translocation of CRT from the endoplasmic reticulum to the cell membrane of tumor cells, concomitantly triggering immunogenic cell death (ICD). In terms of mechanisms, endoplasmic reticulum (ER) stress relievers could impede the ability of PE to induce immunogenicity. This indicates that PE is activated by ER stress, leading to subsequent induction of ICD. Upon analyzing RNA-seq data, it was observed that PE primarily induces programmed cell death in tumors by impeding upstream antioxidant mechanisms. Additionally, it transforms dying tumor cells into vaccines, activating a series of immune responses. CONCLUSIONS: This study observed for the first time that PE-induced CRT exposure on the membrane of cervical cancer cells compensates for the defect of nonimmunogenic cell death inducer CDDP thereby stimulating potent ICD. This ability restores the immunogenicity of CDDP through ER stress induced by the ROS signal. ROS played a role in PE's ability to induce ICD, leading to increased expression of ER stress-related proteins, including ATF3 and IRE-1α. PE exerted anti-cancer effects by increasing the ROS levels, and ROS/ERS signaling may be a potential avenue for cervical cancer treatment. Hence, the synergistic use of PE and CDDP holds potential for enhancing immunochemotherapy in cancer treatment.


Asunto(s)
Calreticulina , Cisplatino , Estrés del Retículo Endoplásmico , Muerte Celular Inmunogénica , Pinellia , Especies Reactivas de Oxígeno , Neoplasias del Cuello Uterino , Cisplatino/farmacología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Femenino , Pinellia/química , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Animales , Extractos Vegetales/farmacología , Proteína HMGB1/metabolismo , Ratones , Línea Celular Tumoral , Ratones Endogámicos BALB C , Células HeLa , Antineoplásicos/farmacología
3.
Exp Cell Res ; 410(1): 112948, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34826423

RESUMEN

Honey-processed Astragalus is a dosage form of Radix Astragalus mixed with honey by a traditional Chinese medicine processing method which improves immune activity. This pharmacological activity of honey-processed Astragalus polysaccharide (HP-APS) might be due to structural changes during the honey roasting process. Previously, we have prepared and characterized HP-APS and preliminarily found its anti-inflammatory effects. However, whether the pharmacodynamic activity of HP-APS induces tumor cell apoptosis and the mechanisms responsible for the immunogenic death (ICD) have not been elucidated. Here, A549, MC38 and B16 cells were used to evaluate the cells viability, apoptosis and cell cycles, respectively. Cellular immunogenic cell death-related molecules calreticulin (CRT), Heat Shock Proteins (HSP)70, major histocompatibility complex I (MHC-I), and co-stimulator molecules CD80/CD86 were determined by flow cytometry. The extracellular ATP release was also detected. B16-OVA and MC38-OVA cells were treated with HP-APS and co-cultivated with OT1 mouse of CD3+T cells for assessment of proliferation, in mice model, and the establishment of C57BL/B6 mouse model bearing B16 cells for assessment of HP-APS the regulation of immune activity in vivo. Our results showed that HP-APS has an inhibitory effect on tumor cell proliferation, which induces tumor cell apoptosis, preventing cells-transforming from G1 phase to S phase in cell cycles. Furthermore, HP-APS could effectively increase the expression of HSP70, CRT, MHC-I, CD86, CD80 and ATP release. T cell proliferation index is significantly improved. CD3 cell proliferation in OT1 mice was significantly increased from the 4th generation to the 5th generation. Moreover, the results have also shown that HP-APS could inhibit tumor growth by increasing immune cell infiltration in the tumor tissues. In the mouse melanoma model with HP-APS treatment, the tumor weight and volume were significantly reduced, and the growth of melanoma was inhibited. CD8+ T is significantly increased. The ratio of CD4+ T and CD8+ T cells numbers are also significantly increased in mouse spleen, but it is less than PD-1 alone treatment separately. Altogether, these findings suggest that HP-APS exerts anti-tumor effects, and that its underlying mechanisms might be associated with the expression of immunogenicity cell death related molecules and the immunomodulatory effects of immune cells.


Asunto(s)
Planta del Astrágalo/química , Medicamentos Herbarios Chinos/farmacología , Muerte Celular Inmunogénica/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Células A549 , Animales , Apoptosis/efectos de los fármacos , Astragalus propinquus/química , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medicamentos Herbarios Chinos/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Miel/análisis , Humanos , Muerte Celular Inmunogénica/inmunología , Inmunomodulación/efectos de los fármacos , Inmunomodulación/inmunología , Activación de Linfocitos/efectos de los fármacos , Melanoma Experimental , Ratones , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Polisacáridos/química , Polisacáridos/farmacología
4.
J Nanobiotechnology ; 19(1): 433, 2021 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-34930269

RESUMEN

BACKGROUND: The construction of a nanoimmune controlled-release system that spatiotemporally recognizes tumor lesions and stimulates the immune system response step by step is one of the most potent cancer treatment strategies for improving the sensitivity of immunotherapy response. RESULTS: Here, a composite nanostimulator (CNS) was constructed for the release of second near-infrared (NIR-II) photothermal-mediated immune agents, thereby achieving spatiotemporally controllable photothermal-synergized immunotherapy. CNS nanoparticles comprise thermosensitive liposomes as an outer shell and are internally loaded with a NIR-II photothermal agent, copper sulfide (CuS), toll-like receptor-9 (TLR-9) agonist, cytosine-phospho-guanine oligodeoxynucleotides, and programmed death-ligand 1 (PD-L1) inhibitors (JQ1). Following NIR-II photoirradiation, CuS enabled the rapid elevation of localized temperature, achieving tumor ablation and induction of immunogenic cell death (ICD) as well as disruption of the lipid shell, enabling the precise release of two immune-therapeutical drugs in the tumor region. Combining ICD, TLR-9 stimulation, and inhibited expression of PD-L1 allows the subsequent enhancement of dendritic cell maturation and increases infiltration of cytotoxic T lymphocytes, facilitating regional antitumor immune responses. CONCLUSION: CNS nanoparticle-mediated photothermal-synergized immunotherapy efficiently suppressed the growth of primary and distant tumors in two mouse models and prevented pulmonary metastasis. This study thus provides a novel sight into photo-controllably safe and efficient immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Rayos Infrarrojos , Nanopartículas/química , Neoplasias/terapia , Fototerapia/métodos , Animales , Azepinas/química , Azepinas/farmacología , Azepinas/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Cobre/química , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Muerte Celular Inmunogénica/efectos de los fármacos , Verde de Indocianina/química , Verde de Indocianina/uso terapéutico , Liposomas/química , Ratones , Ratones Endogámicos C57BL , Neoplasias/patología , Receptor Toll-Like 9/metabolismo , Trasplante Heterólogo , Triazoles/química , Triazoles/farmacología , Triazoles/uso terapéutico
5.
J Nanobiotechnology ; 19(1): 455, 2021 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-34963499

RESUMEN

BACKGROUND: The complex tumor microenvironment and non-targeting drugs limit the efficacy of clinical tumor therapy. For ensuring the accurate delivery and maximal effects of anticancer drugs, it is important to develop innovative drug delivery system based on nano-strategies. RESULT: In this study, an intracellular acidity-responsive polymeric metal organic framework nanoparticle (denoted as DIMP) has been constructed, which can co-deliver the chemotherapy agent of doxorubicin (DOX) and phototherapy agent of indocyanine green (ICG) for breast carcinoma theranostics. Specifically, DIMP possesses a suitable and stable nanometer size and can respond to the acidic microenvironment in cells, thus precisely delivering drugs into target tumor sites and igniting the biological reactions towards cell apoptosis. Following in vivo and in vitro results showed that DIMP could be effectively accumulated in tumor sites and induced powerful immunogenic cell death (ICD) effect. CONCLUSION: The designed DIMP displayed its effectiveness in combined photo-chemotherapy with auxiliary of ICD effect under a multimodal imaging monitor. Thus, the present MOF-based strategy may offer a potential paradigm for designing drug-delivery system for image-guided synergistic tumor therapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Muerte Celular Inmunogénica/efectos de los fármacos , Estructuras Metalorgánicas/administración & dosificación , Nanopartículas/administración & dosificación , Microambiente Tumoral/efectos de los fármacos , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Terapia Combinada , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Femenino , Verde de Indocianina/administración & dosificación , Verde de Indocianina/química , Verde de Indocianina/farmacología , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacología , Ratones , Imagen Multimodal , Nanopartículas/química , Fototerapia
6.
Nat Commun ; 12(1): 4299, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34262038

RESUMEN

Radiofrequency ablation (RFA) is clinically adopted to destruct solid tumors, but is often incapable of completely ablating large tumors and those with multiple metastatic sites. Here we develop a CaCO3-assisted double emulsion method to encapsulate lipoxidase and hemin with poly(lactic-co-glycolic acid) (PLGA) to enhance RFA. We show the HLCaP nanoreactors (NRs) with pH-dependent catalytic capacity can continuously produce cytotoxic lipid radicals via the lipid peroxidation chain reaction using cancer cell debris as the fuel. Upon being fixed inside the residual tumors post RFA, HLCaP NRs exhibit a suppression effect on residual tumors in mice and rabbits by triggering ferroptosis. Moreover, treatment with HLCaP NRs post RFA can prime antitumor immunity to effectively suppress the growth of both residual and metastatic tumors, also in combination with immune checkpoint blockade. This work highlights that tumor-debris-fueled nanoreactors can benefit RFA by inhibiting tumor recurrence and preventing tumor metastasis.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Nanomedicina/métodos , Neoplasias/terapia , Ablación por Radiofrecuencia , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/farmacología , Animales , Carbonato de Calcio/química , Carbonato de Calcio/uso terapéutico , Catálisis , Línea Celular Tumoral , Terapia Combinada , Ferroptosis/efectos de los fármacos , Hemina/química , Hemina/uso terapéutico , Humanos , Concentración de Iones de Hidrógeno , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Muerte Celular Inmunogénica/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Lipooxigenasa/química , Lipooxigenasa/uso terapéutico , Ratones , Metástasis de la Neoplasia , Neoplasia Residual , Neoplasias/inmunología , Neoplasias/patología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/uso terapéutico , Conejos
7.
Mar Drugs ; 19(2)2021 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-33499163

RESUMEN

Marine plants have become an inexhaustible reservoir of new phytopharmaceuticals for cancer treatment. We demonstrate in vitro/in vivo antitumor efficacy of a standardized polyphenol extract from the marine angiosperm Thalassia testudinum (TTE) in colon tumor cell lines (RKO, SW480, and CT26) and a syngeneic allograft murine colorectal cancer model. MTT assays revealed a dose-dependent decrease of cell viability of RKO, CT26, and SW480 cells upon TTE treatment with IC50 values of, respectively, 175, 115, and 60 µg/mL. Furthermore, TTE significantly prevented basal and bFGF-induced angiogenesis in the chicken chorioallantoic membrane angiogenesis assay. In addition, TTE suppressed bFGF-induced migration of endothelial cells in a wound closure assay. Finally, TTE treatment abrogated CT26 colorectal cancer growth and increased overall organism survival in a syngeneic murine allograft model. Corresponding transcriptome profiling and pathway analysis allowed for the identification of the mechanism of action for the antitumor effects of TTE. In line with our in vitro/in vivo results, TTE treatment triggers ATF4-P53-NFκB specific gene expression and autophagy stress pathways. This results in suppression of colon cancer cell growth, cell motility, and angiogenesis pathways in vitro and in addition promotes antitumor immunogenic cell death in vivo.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Hydrocharitaceae , Muerte Celular Inmunogénica/efectos de los fármacos , Neovascularización Patológica/patología , Extractos Vegetales/uso terapéutico , Animales , Antineoplásicos Fitogénicos/aislamiento & purificación , Antineoplásicos Fitogénicos/farmacología , Autofagia/efectos de los fármacos , Autofagia/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Humanos , Hydrocharitaceae/química , Muerte Celular Inmunogénica/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
8.
Angew Chem Int Ed Engl ; 59(49): 22202-22209, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32841465

RESUMEN

A silver-ion-coupled black phosphorus (BP) vesicle (BP Ve-Ag+ ) with a second near infrared (NIR-II) window photoacoustic (PA) imaging capability was firstly constructed to maximize the potential of BP quantum dot (QD) in deeper bioimaging and diversified therapy. The embedded Ag+ could improve the relatively large band gap of BP QD via intense charge coupling based on theoretical simulation results, subsequently leading to the enhanced optical absorption capability, accompanied with the occurrence of the strong NIR-II PA signal. Guiding by NIR-II PA bioimaging, the hidden Ag+ could be precisely released with the disassembly of Ve during photodynamic therapy process and captured by macrophages located in lesion region for arousing synergistic cancer photodynamic/Ag+ immunotherapy. BP Ve-Ag+ can contrapuntally kill pathogenic bacteria and accelerate wound healing monitored by NIR-II PA imaging.


Asunto(s)
Antineoplásicos/farmacología , Fósforo/farmacología , Técnicas Fotoacústicas , Fotoquimioterapia , Plata/farmacología , Cicatrización de Heridas/efectos de los fármacos , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Teoría Funcional de la Densidad , Ensayos de Selección de Medicamentos Antitumorales , Muerte Celular Inmunogénica/efectos de los fármacos , Muerte Celular Inmunogénica/inmunología , Rayos Infrarrojos , Ratones , Tamaño de la Partícula , Fósforo/química , Puntos Cuánticos/química , Células RAW 264.7 , Plata/química , Propiedades de Superficie , Linfocitos T Citotóxicos/inmunología
10.
ACS Nano ; 13(11): 12638-12652, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31625721

RESUMEN

Photodynamic therapy (PDT) is a clinical cancer treatment modality based on the induction of therapeutic reactive oxygen species (ROS), which can trigger immunogenic cell death (ICD). With the aim of simultaneously improving both PDT-mediated intracellular ROS production and ICD levels, we designed a serum albumin (SA)-coated boehmite ("B"; aluminum hydroxide oxide) organic-inorganic scaffold that could be loaded with chlorin e6 (Ce6), a photosensitizer, and a honey bee venom melittin (MLT) peptide, denoted Ce6/MLT@SAB. Ce6/MLT@SAB was anchored by a boehmite nanorod structure and exhibited particle size of approximately 180 nm. Ce6/MLT@SAB could significantly reduce hemolysis relative to that of free MLT, while providing MLT-enhanced PDT antitumor effects in vitro. Compared with Ce6@SAB, Ce6/MLT@SAB improved Ce6 penetration of cancer cells both in vitro and in vivo, thereby providing enhanced intracellular ROS generation with 660 nm light treatment. Following phototreatment, Ce6/MLT@SAB-treated cells displayed significantly improved levels of ICD and abilities to activate dendritic cells. In the absence of laser irradiation, multidose injection of Ce6/MLT@SAB could delay the growth of subcutaneous murine tumors by more than 60%, compared to controls. When combined with laser irradiation, a single injection and phototreatment with Ce6/MLT@SAB eradicated one-third of subcutaneous tumors in treated mice. The addition of an immune checkpoint blockade to Ce6/MLT@SAB phototreatment further augmented antitumor effects, generating increased numbers of CD4+ and CD8+ T cells in tumors with concomitant reduction of myeloid-derived suppressor cells.


Asunto(s)
Antineoplásicos , Inmunoterapia/métodos , Meliteno , Nanopartículas/química , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Femenino , Muerte Celular Inmunogénica/efectos de los fármacos , Meliteno/química , Meliteno/farmacocinética , Meliteno/farmacología , Ratones , Ratones Endogámicos BALB C , Neoplasias Experimentales/patología , Fármacos Fotosensibilizantes/química , Fármacos Fotosensibilizantes/farmacocinética , Fármacos Fotosensibilizantes/farmacología
11.
Nano Lett ; 19(11): 8333-8341, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31657935

RESUMEN

A group of chemotherapeutic drugs has gained increasing interest in cancer immunotherapy due to the potential to induce immunogenic cell death (ICD). A critical challenge in using the ICD inducers in cancer immunotherapy is the immunotoxicity accompanying their antiproliferative effects. To alleviate this, a nanocapsule formulation of carfilzomib (CFZ), an ICD-inducing proteasome inhibitor, was developed using interfacial supramolecular assembly of tannic acid (TA) and iron, supplemented with albumin coating. The albumin-coated CFZ nanocapsules (CFZ-pTA-alb) attenuated CFZ release, reducing toxicity to immune cells. Moreover, due to the adhesive nature of the TA assembly, CFZ-pTA-alb served as a reservoir of damage-associated molecular patterns released from dying tumor cells to activate dendritic cells. Upon intratumoral administration, CFZ-pTA-alb prolonged tumor retention of CFZ and showed consistently greater antitumor effects than cyclodextrin-solubilized CFZ (CFZ-CD) in B16F10 and CT26 tumor models. Unlike CFZ-CD, the locally injected CFZ-pTA-alb protected or enhanced CD8+ T cell population in tumors, helped develop splenocytes with tumor-specific interferon-γ response, and delayed tumor development on the contralateral side in immunocompetent mice (but not in athymic nude mice), supporting that CFZ-pTA-alb contributed to activating antitumor immunity. This study demonstrates that sustained delivery of ICD inducers by TA-based nanocapsules is an effective way of translating local ICD induction to systemic antitumor immunity.


Asunto(s)
Antineoplásicos/administración & dosificación , Nanocápsulas/química , Neoplasias/tratamiento farmacológico , Oligopéptidos/administración & dosificación , Taninos/química , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Preparaciones de Acción Retardada/química , Humanos , Inmunidad/efectos de los fármacos , Muerte Celular Inmunogénica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Oligopéptidos/uso terapéutico
12.
Acc Chem Res ; 52(8): 2332-2339, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31335117

RESUMEN

This Account is the first comprehensive review article on the newly developed, photochemistry-based cancer therapy near-infrared (NIR) photoimmunotherapy (PIT). NIR-PIT is a molecularly targeted phototherapy for cancer that is based on injecting a conjugate of a near-infrared, water-soluble, silicon-phthalocyanine derivative, IRdye700DX (IR700), and a monoclonal antibody (mAb) that targets an expressed antigen on the cancer cell surface. Subsequent local exposure to NIR light turns on this photochemical "death" switch, resulting in the rapid and highly selective immunogenic cell death (ICD) of targeted cancer cells. ICD occurs as early as 1 min after exposure to NIR light and results in irreversible morphologic changes only in target-expressing cells based on the newly discovered photoinduced ligand release reaction that induces physical changes on conjugated antibody/antigen complex resulting in functional damage on cell membrane. Meanwhile, immediately adjacent receptor-negative cells are totally unharmed. Because of its highly targeted nature, NIR-PIT carries few side effects and healing is rapid. Evaluation of the tumor microenvironment reveals that ICD induced by NIR-PIT results in rapid maturation of immature dendritic cells adjacent to dying cancer cells initiating a host anticancer immune response, resulting in repriming of polyclonal CD8+T cells against various released cancer antigens, which amplifies the therapeutic effect of NIR-PIT. NIR-PIT can target and treat virtually any cell surface antigens including cancer stem cell markers, that is, CD44 and CD133. A first-in-human phase 1/2 clinical trial of NIR-PIT using cetuximab-IR700 (RM1929) targeting EGFR in inoperable recurrent head and neck cancer patients successfully concluded in 2017 and led to "fast tracking" by the FDA and a phase 3 trial ( https://clinicaltrials.gov/ct2/show/NCT03769506 ) that is currently underway in 3 countries in Asia, US/Canada, and 4 countries in EU. The next step for NIR-PIT is to further exploit the immune response. Preclinical research in animals with intact immune systems has shown that NIT-PIT targeting of immunosuppressor cells within the tumor, such as regulatory T-cells, can further enhance tumor-cell-selective systemic host-immunity leading to significant responses in distant metastatic tumors, which are not treated with light. By combining cancer-targeting NIR-PIT and immune-activating NIR-PIT or other cancer immunotherapies, NIR-PIT of a local tumor, could lead to responses in distant metastases and may also inhibit recurrences due to activation of systemic anticancer immunity and long-term immune memory without the systemic autoimmune adverse effects often associated with immune checkpoint inhibitors. Furthermore, NIR-PIT also enhances nanodrug delivery into tumors up to 24-fold superior to untreated tumors with conventional EPR effects by intensively damaging cancer cells behind tumor vessels. We conclude by describing future advances in this novel photochemical cancer therapy that are likely to further enhance the efficacy of NIR-PIT.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Neoplasias/terapia , Fármacos Fotosensibilizantes/uso terapéutico , Animales , Antineoplásicos/efectos de la radiación , Ensayos Clínicos como Asunto , Células Dendríticas/efectos de los fármacos , Humanos , Inmunoconjugados/efectos de la radiación , Muerte Celular Inmunogénica/efectos de los fármacos , Inmunoterapia/métodos , Rayos Infrarrojos , Ratones , Fármacos Fotosensibilizantes/efectos de la radiación , Fototerapia/métodos
13.
Angew Chem Int Ed Engl ; 58(3): 670-680, 2019 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-30016571

RESUMEN

Cancer immunotherapies that train or stimulate the inherent immunological systems to recognize, attack, and eradicate tumor cells with minimal damage to healthy cells have demonstrated promising clinical responses in recent years. However, most of these immunotherapeutic strategies only benefit a small subset of patients and cause systemic autoimmune side effects in some patients. Immunogenic cell death (ICD)-inducing modalities not only directly kill cancer cells but also induce antitumor immune responses against a broad spectrum of solid tumors. Such strategies for generating vaccine-like functions could be used to stimulate a "cold" tumor microenvironment to become an immunogenic, "hot" tumor microenvironment, working in synergy with immunotherapies to increase patient response rates and lead to successful treatment outcomes. This Minireview will focus on nanoparticle-based treatment modalities that can induce and enhance ICD to potentiate cancer immunotherapy.


Asunto(s)
Muerte Celular Inmunogénica , Inmunoterapia/métodos , Nanopartículas/uso terapéutico , Neoplasias/terapia , Animales , Antineoplásicos/uso terapéutico , Humanos , Hipertermia Inducida/métodos , Muerte Celular Inmunogénica/efectos de los fármacos , Nanomedicina/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/radioterapia , Fotoquimioterapia/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA