Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 29: 235-71, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21219185

RESUMEN

The immune system can identify and destroy nascent tumor cells in a process termed cancer immunosurveillance, which functions as an important defense against cancer. Recently, data obtained from numerous investigations in mouse models of cancer and in humans with cancer offer compelling evidence that particular innate and adaptive immune cell types, effector molecules, and pathways can sometimes collectively function as extrinsic tumor-suppressor mechanisms. However, the immune system can also promote tumor progression. Together, the dual host-protective and tumor-promoting actions of immunity are referred to as cancer immunoediting. In this review, we discuss the current experimental and human clinical data supporting a cancer immunoediting process that provide the fundamental basis for further study of immunity to cancer and for the rational design of immunotherapies against cancer.


Asunto(s)
Neoplasias/inmunología , Inmunidad Adaptativa , Animales , Humanos , Inmunidad Innata , Vigilancia Inmunológica , Neoplasias/fisiopatología , Neoplasias/terapia
2.
Proc Natl Acad Sci U S A ; 116(50): 25229-25235, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31767744

RESUMEN

Responses of solid tumors to chimeric antigen receptor (CAR) T cell therapy are often minimal. This is potentially due to a lack of sustained activation and proliferation of CAR T cells when encountering antigen in a profoundly immunosuppressive tumor microenvironment. In this study, we investigate if inducing an interaction between CAR T cells and antigen-presenting cells (APCs) in lymphoid tissue, away from an immunosuppressive microenvironment, could enhance solid-tumor responses. We combined CAR T cell transfer with the bacterial enterotoxin staphylococcal enterotoxin-B (SEB), which naturally links a proportion of T cell receptor (TCR) Vß subtypes to MHC-II, present on APCs. CAR T cell proliferation and function was significantly enhanced by SEB. Solid tumor-growth inhibition in mice was increased when CAR T cells were administered in combination with SEB. CAR T cell expansion in lymphoid tissue was demonstrated, and inhibition of lymphocyte egress from lymph nodes using FTY720 abrogated the benefit of SEB. We also demonstrate that a bispecific antibody, targeting a c-Myc tag on CAR T cells and cluster of differentiation 40 (CD40), could also enhance CAR T cell activity and mediate increased antitumor activity of CAR T cells. These model systems serve as proof-of-principle that facilitating the interaction of CAR T cells with APCs can enhance their ability to mediate antitumor activity.


Asunto(s)
Enterotoxinas/farmacología , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Antígenos CD40/inmunología , Proliferación Celular/efectos de los fármacos , Humanos , Inmunoterapia Adoptiva , Ratones , Ratones Endogámicos C57BL , Neoplasias/genética , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/citología
3.
Semin Immunol ; 28(1): 64-72, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26611350

RESUMEN

The frontiers of cancer immunotherapy are extending in terms of both the range of cancer types that can potentially be targeted and the types of therapeutics that are in clinical development. The use of adoptive cellular therapy (ACT) and its derivative, chimeric antigen receptor (CAR) T cells, is currently limited to hematological malignancies and immunogenic cancers such as melanoma and renal cell carcinoma. Although ACT utilizing ex vivo expanded tumor-infiltrating lymphocytes (TIL) or engineered CAR/TCR T cells have undergone clinical trials for other solid cancers, their efficacy to date has been limited. This may be due, in part, to the immunosuppressive nature of the tumor microenvironment. The development of novel combination approaches which target the immunosuppressive network engineered by tumors has raised the possibility of using ACT for a broader range of cancers. This review summarizes the potential of such strategies and outlines the clinical relevance of these observations.


Asunto(s)
Antineoplásicos/uso terapéutico , Vacunas contra el Cáncer/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Supervivencia Celular , Terapia Combinada , Receptores Coestimuladores e Inhibidores de Linfocitos T/metabolismo , Humanos , Terapia de Inmunosupresión , Activación de Linfocitos , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/trasplante , Microambiente Tumoral
4.
Immunol Cell Biol ; 95(4): 356-363, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28003642

RESUMEN

The potential for immunotherapy as a treatment option for cancer is clear from remarkable responses of some leukemia patients to adoptive cell transfer using autologous T cells genetically modified to express chimeric antigen receptors (CARs). However, the vast majority of cancers, in particular the more common solid cancers, such as those of the breast, colon and lung, fail to respond significantly to infusions of CAR T cells. Solid cancers present some formidable barriers to adoptive cell transfer, including suppression of T-cell function and inhibition of T-cell localization. In this review, we discuss the current state of CAR T-cell therapy in solid cancers, the variety of concepts being investigated to overcome these barriers as well as approaches aimed at increasing the specificity and safety of adoptive cell transfer.


Asunto(s)
Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/inmunología , Neoplasias del Colon/terapia , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/terapia , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Neoplasias de la Mama/inmunología , Movimiento Celular , Neoplasias del Colon/inmunología , Ingeniería Genética , Humanos , Tolerancia Inmunológica , Neoplasias Pulmonares/inmunología , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/genética , Linfocitos T/trasplante , Escape del Tumor
5.
Nat Rev Immunol ; 5(12): 928-40, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322746

RESUMEN

Immunotherapy is receiving much attention as a means of treating cancer, but complete, durable responses remain rare for most malignancies. The natural immune system seems to have limitations and deficiencies that might affect its ability to control malignant disease. An alternative to relying on endogenous components in the immune repertoire is to generate lymphocytes with abilities that are greater than those of natural T cells, through genetic modification to produce 'supernatural' T cells. This Review describes how such T cells can circumvent many of the barriers that are inherent in the tumour microenvironment while optimizing T-cell specificity, activation, homing and antitumour function.


Asunto(s)
Vacunas contra el Cáncer , Terapia Genética/métodos , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Quimera , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología
6.
Mol Ther ; 22(1): 18-27, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24048441

RESUMEN

The tumor microenvironment can promote tumor growth and reduce treatment efficacy. Tumors can occur in many sites in the body, but how surrounding normal tissues at different anatomical sites affect tumor microenvironments and their subsequent response to therapy is not known.We demonstrated that tumors from renal, colon, or prostate cell lines in orthotopic locations responded to immunotherapy consisting of three agonist antibodies, termed Tri-mAb, to a much lesser extent than the same tumor type located subcutaneously. A tissue-specific response to Tri-mAb was confirmed by ex vivo separation of subcutaneous (SC) or orthotopic tumor cells from stromal cells, followed by reinjection of tumor cells into the opposite site. Compared with SC tumors, orthotopic tumors had a microenvironment associated with a type 2 immune response, related to immunosuppression, and an involvement of alternatively activated macrophages in the kidney model. Orthotopic kidney tumors were more highly vascularized than SC tumors. Neutralizing the macrophage- and Th2-associated molecules chemokine (C-C motif) ligand 2 or interleukin-13 led to a significantly improved therapeutic effect. This study highlights the importance of the tissue of implantation in sculpting the tumor microenvironment. These are important fundamental issues in tumor biology and crucial factors to consider in the design of experimental models and treatment strategies.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD40/antagonistas & inhibidores , Antígenos CD40/inmunología , Línea Celular Tumoral , Quimiocina CCL2/inmunología , Neoplasias del Colon/inmunología , Modelos Animales de Enfermedad , Expresión Génica , Interleucina-13/inmunología , Neoplasias Renales/inmunología , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Neoplasias/mortalidad , Neoplasias/terapia , Neovascularización Patológica/inmunología , Especificidad de Órganos/inmunología , Próstata/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/antagonistas & inhibidores , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Resultado del Tratamiento , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
7.
Cancer Immunol Immunother ; 63(9): 869-76, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25063364

RESUMEN

Forkhead box P3 (Foxp3) is an important transcription factor that belongs to the forkhead/winged-helix family of transcriptional regulators. Foxp3 has been extensively studied over the past 13 years as a master regulator of transcription in a specific T-cell type, CD4(+) regulatory T cells (Treg), both in humans and in mice. Compelling data characterize Foxp3 as critically important and necessary for the development and the differentiation of Treg. It has been considered initially as the only specific marker for Treg. However, recent work has proposed that Foxp3 can be expressed by other types of lymphoid cells or myeloid cells and also by some non-hematopoietic cells such as epithelial cells. It remains controversial about the expression of Foxp3 in cells other than Treg, but understanding the potential expression and function of this master regulator in different cell subsets could have a wide range of implications for immune tolerance and several pathologies including autoimmune disorders and immune responses to cancer.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Linfocitos T Reguladores/metabolismo , Animales , Linaje de la Célula , Factores de Transcripción Forkhead/inmunología , Humanos , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
8.
Mol Ther ; 21(11): 2122-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23831595

RESUMEN

In a phase I study of autologous chimeric antigen receptor (CAR) anti-LeY T-cell therapy of acute myeloid leukemia (AML), we examined the safety and postinfusion persistence of adoptively transferred T cells. Following fludarabine-containing preconditioning, four patients received up to 1.3 × 109 total T cells, of which 14-38% expressed the CAR. Grade 3 or 4 toxicity was not observed. One patient achieved a cytogenetic remission whereas another with active leukemia had a reduction in peripheral blood (PB) blasts and a third showed a protracted remission. Using an aliquot of In111-labeled CAR T cells, we demonstrated trafficking to the bone marrow (BM) in those patients with the greatest clinical benefit. Furthermore, in a patient with leukemia cutis, CAR T cells infiltrated proven sites of disease. Serial PCR of PB and BM for the LeY transgene demonstrated that infused CAR T cells persisted for up to 10 months. Our study supports the feasibility and safety of CAR-T-cell therapy in high-risk AML, and demonstrates durable in vivo persistence.


Asunto(s)
Inmunoterapia Adoptiva , Leucemia Mieloide Aguda/terapia , Antígenos del Grupo Sanguíneo de Lewis/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Anciano , Médula Ósea/inmunología , Femenino , Humanos , Leucemia Mieloide Aguda/inmunología , Masculino , Persona de Mediana Edad , Inducción de Remisión , Acondicionamiento Pretrasplante , Vidarabina/análogos & derivados , Vidarabina/uso terapéutico
9.
Blood ; 118(3): 499-509, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21531979

RESUMEN

In this age of promise of new therapies for cancer, immunotherapy is emerging as an exciting treatment option for patients. Vaccines and cytokines are being tested extensively in clinical trials, and strategies using monoclonal antibodies and cell transfer are mediating dramatic regression of tumors in patients with certain malignancies. However, although initially advocated as being more specific for cancer and having fewer side effects than conventional therapies, it is becoming increasingly clear that many immunotherapies can lead to immune reactions against normal tissues. Immunotoxicities resulting from treatment can range from relatively minor conditions, such as skin depigmentation, to severe toxicities against crucial organ systems, such as liver, bowel, and lung. Treatment-related toxicity has correlated with better responses in some cases, and it is probable that serious adverse events from immune-mediated reactions will increase in frequency and severity as immunotherapeutic approaches become more effective. This review introduces immunotherapeutic approaches to cancer treatment, provides details of toxicities arising from therapy, and discusses future potential ways to avoid or circumvent these side effects.


Asunto(s)
Autoinmunidad/inmunología , Inmunoterapia/efectos adversos , Neoplasias/inmunología , Neoplasias/terapia , Humanos , Inmunoterapia/métodos , Inmunoterapia/tendencias
10.
Cancers (Basel) ; 15(4)2023 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-36831663

RESUMEN

This Special Issue reminds us that, although incredible developments have occurred in the field of cancer immunotherapy, there is still plenty of room for improvement [...].

11.
Sci Transl Med ; 15(690): eabk1900, 2023 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-37018415

RESUMEN

Patients who receive chimeric antigen receptor (CAR)-T cells that are enriched in memory T cells exhibit better disease control as a result of increased expansion and persistence of the CAR-T cells. Human memory T cells include stem-like CD8+ memory T cell progenitors that can become either functional stem-like T (TSTEM) cells or dysfunctional T progenitor exhausted (TPEX) cells. To that end, we demonstrated that TSTEM cells were less abundant in infused CAR-T cell products in a phase 1 clinical trial testing Lewis Y-CAR-T cells (NCT03851146), and the infused CAR-T cells displayed poor persistence in patients. To address this issue, we developed a production protocol to generate TSTEM-like CAR-T cells enriched for expression of genes in cell replication pathways. Compared with conventional CAR-T cells, TSTEM-like CAR-T cells had enhanced proliferative capacity and increased cytokine secretion after CAR stimulation, including after chronic CAR stimulation in vitro. These responses were dependent on the presence of CD4+ T cells during TSTEM-like CAR-T cell production. Adoptive transfer of TSTEM-like CAR-T cells induced better control of established tumors and resistance to tumor rechallenge in preclinical models. These more favorable outcomes were associated with increased persistence of TSTEM-like CAR-T cells and an increased memory T cell pool. Last, TSTEM-like CAR-T cells and anti-programmed cell death protein 1 (PD-1) treatment eradicated established tumors, and this was associated with increased tumor-infiltrating CD8+CAR+ T cells producing interferon-γ. In conclusion, our CAR-T cell protocol generated TSTEM-like CAR-T cells with enhanced therapeutic efficacy, resulting in increased proliferative capacity and persistence in vivo.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Humanos , Inmunoterapia Adoptiva/métodos , Linfocitos T , Citocinas/metabolismo , Células Madre/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo
12.
J Immunol ; 185(1): 532-41, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20505139

RESUMEN

The selective targeting of the tumor-associated death-inducing receptors DR4 and DR5 with agonistic mAbs has demonstrated preclinical and clinical antitumor activity. However, the cellular and molecular mechanisms contributing to this efficacy remain poorly understood. In this study, using the first described C57BL/6 (B6) TRAIL-sensitive experimental tumor models, we have characterized the innate and adaptive immune components involved in the primary rejection phase of an anti-mouse DR5 (mDR5) mAb, MD5-1 in established MC38 colon adenocarcinomas. FcR mediated cross-linking of MD5-1 significantly inhibited the growth of MC38 colon adenocarcinomas through the induction of TRAIL-R-dependent tumor cell apoptosis. The loss of host DR5, TRAIL, perforin, FasL, or TNF did not compromise anti-DR5 therapy in vivo. By contrast, anti-DR5 therapy was completely abrogated in mice deficient of B cells or CD11c(+) dendritic cells (DCs), providing the first direct evidence that these cells play a critical role. Importantly, the requirement for an intact B cell compartment for optimal anti-DR5 antitumor efficacy was also observed in established AT-3 mammary tumors. Interestingly, MD5-1-mediated apoptosis as measured by early TUNEL activity was completely lost in B cell-deficient microMT mice, but intact in mice deficient in CD11c(+) DCs. Overall, these data show that Ab-mediated targeting of DR5 triggers tumor cell apoptosis in established tumors in a B cell-dependent manner and that CD11c(+) DCs make a critical downstream contribution to anti-DR5 antitumor activity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Subgrupos de Linfocitos B/inmunología , Antígeno CD11c/fisiología , Células Dendríticas/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Adenocarcinoma/terapia , Animales , Anticuerpos Monoclonales/toxicidad , Subgrupos de Linfocitos B/metabolismo , Antígeno CD11c/biosíntesis , Línea Celular , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/terapia , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/terapia , Mastocitoma/inmunología , Mastocitoma/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/deficiencia , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética
13.
Cancer Immunol Immunother ; 60(5): 671-83, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21327636

RESUMEN

Toll-like receptor (TLR) agonists can trigger broad inflammatory responses that elicit rapid innate immunity and promote the activities of lymphocytes, which can potentially enhance adoptive immunotherapy in the tumor-bearing setting. In the present study, we found that Polyinosinic:Polycytidylic Acid [Poly(I:C)] and CpG oligodeoxynucleotide 1826 [CpG], agonists for TLR 3 and 9, respectively, potently activated adoptively transferred T cells against a murine model of established melanoma. Intratumoral injection of Poly(I:C) and CpG, combined with systemic transfer of activated pmel-1 T cells, specific for gp100(25-33), led to enhanced survival and eradication of 9-day established subcutaneous B16F10 melanomas in a proportion of mice. A series of survival studies in knockout mice supported a key mechanistic pathway, whereby TLR agonists acted via host cells to enhance IFN-γ production by adoptively transferred T cells. IFN-γ, in turn, enhanced the immunogenicity of the B16F10 melanoma line, leading to increased killing by adoptively transferred T cells. Thus, this combination approach counteracted tumor escape from immunotherapy via downregulation of immunogenicity. In conclusion, TLR agonists may represent advanced adjuvants within the setting of adoptive T-cell immunotherapy of cancer and hold promise as a safe means of enhancing this approach within the clinic.


Asunto(s)
Inmunoterapia Adoptiva , Melanoma Experimental/terapia , Oligodesoxirribonucleótidos/uso terapéutico , Poli I-C/uso terapéutico , Linfocitos T/inmunología , Receptores Toll-Like/agonistas , Adyuvantes Inmunológicos/administración & dosificación , Animales , Línea Celular Tumoral , Células Dendríticas/inmunología , Citometría de Flujo , Inflamación , Interferón gamma/biosíntesis , Activación de Linfocitos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Toll-Like/inmunología
14.
Immunol Cell Biol ; 89(2): 216-24, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20567250

RESUMEN

Natural killer (NK) cells are potent immune effector cells that can respond to infection and cancer, as well as allowing maternal adaptation to pregnancy. In response to malignant transformation or pathogenic invasion, NK cells can secrete cytokine and may be directly cytolytic, as well as exerting effects indirectly through other cells of the immune system. To recognize and respond to inflamed or infected tissues, NK cells express a variety of activating and inhibitory receptors including NKG2D, Ly49 or KIR, CD94-NKG2 heterodimers and natural cytotoxicity receptors, as well as co-stimulatory receptors. These receptors recognize cellular stress ligands as well as major histocompatibility complex class I and related molecules, which can lead to NK cell responses. Importantly, NK cells must remain tolerant of healthy tissue, and some of these receptors can also prevent activation of NK cells. In this review, we describe the expression of prominent NK cell receptors, as well as expression of their ligands and their role in immune responses. In addition, we describe the main signaling pathways used by NK cell receptors. Although we now appreciate that NK cell biology is more complicated than first thought, there are still facets of their biology that remain unclear. These will be highlighted and discussed in this review.


Asunto(s)
Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Receptores de Células Asesinas Naturales/inmunología , Animales , Citotoxicidad Inmunológica , Humanos , Multimerización de Proteína
15.
Immunother Adv ; 1(1): ltab016, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35919743

RESUMEN

Co-stimulation is a fundamental component of T cell biology and plays a key role in determining the quality of T cell proliferation, differentiation, and memory formation. T cell-based immunotherapies, such as chimeric antigen receptor (CAR) T cell immunotherapy, are no exception. Solid tumours have largely been refractory to CAR T cell therapy owing to an immunosuppressive microenvironment which limits CAR T cell persistence and effector function. In order to eradicate solid cancers, increasingly sophisticated strategies are being developed to deliver these vital co-stimulatory signals to CAR T cells, often specifically within the tumour microenvironment. These include designing novel co-stimulatory domains within the CAR or other synthetic receptors, arming CAR T cells with cytokines or using CAR T cells in combination with agonist antibodies. This review discusses the evolving role of co-stimulation in CAR T cell therapies and the strategies employed to target co-stimulatory pathways in CAR T cells, with a view to improve responses in solid tumours.

16.
FEBS J ; 288(1): 81-90, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32248616

RESUMEN

Cancer tissue is not homogenous, and individual metastases at different anatomical locations can differ from the primary tumor and from one another in both their morphology and cellular composition, even within an individual patient. Tumors are composed of cancer cells and a range of other cell types, which, together with a variety of secreted molecules, collectively comprise the tumor microenvironment (TME). Cells of the TME can communicate with each other and with distant tissues in a form of molecular cross-talk to influence their growth and function. Cross-talk between cancer cells and local immune cells is well described and can lead to the induction of local immunosuppression. Recently, it has become apparent that tumors located remotely from each other, can engage in cross-talk that can influence their responsiveness to various therapies, including immunotherapy. In this article, we review studies that describe how tumors systemically communicate with distant tissues through motile cells, extracellular vesicles, and secreted molecules that can affect their function. In addition, we summarize evidence from mouse studies and the clinic that indicate an ability of some tumors to influence the progression and therapeutic responses of other tumors in different anatomical locations.


Asunto(s)
Vesículas Extracelulares/inmunología , Proteínas de Neoplasias/genética , Neoplasias/inmunología , Células Neoplásicas Circulantes/inmunología , Células Madre Neoplásicas/inmunología , Animales , Comunicación Celular , Vesículas Extracelulares/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Inmunoterapia/métodos , Ratones , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/patología , Metástasis de la Neoplasia , Proteínas de Neoplasias/inmunología , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Células Neoplásicas Circulantes/patología , Células Madre Neoplásicas/patología , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
17.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34035114

RESUMEN

Rapid advances in immunotherapy have identified adoptive cell transfer as one of the most promising approaches for the treatment of cancers. Large numbers of cancer reactive T lymphocytes can be generated ex vivo from patient blood by genetic modification to express chimeric antigen receptors (CAR) specific for tumor-associated antigens. CAR T cells can respond strongly against cancer cells, and adoptive transferred CAR T cells can induce dramatic responses against certain types of cancers. The ability of T cells to respond against disease depends on their ability to localize to sites, persist and exert functions, often in an immunosuppressive microenvironment, and these abilities are reflected in their phenotypes. There is currently intense interest in generating CAR T cells possessing the ideal phenotypes to confer optimal antitumor activity. In this article, we review T cell phenotypes for trafficking, persistence and function, and discuss how culture conditions and genetic makeups can be manipulated to achieve the ideal phenotypes for antitumor activities.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/trasplante , Animales , Diferenciación Celular , Proliferación Celular , Autorrenovación de las Células , Citotoxicidad Inmunológica , Humanos , Memoria Inmunológica , Activación de Linfocitos , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Resultado del Tratamiento
18.
Ther Adv Vaccines Immunother ; 9: 25151355211017119, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34159293

RESUMEN

Background: Tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapies have demonstrated promising, though limited, efficacy against melanoma. Methods: We designed a model system to explore the efficacy of dual specific T cells derived from melanoma patient TILs by transduction with a Her2-specific CAR. Results: Metastatic melanoma cells in our biobank constitutively expressed Her2 antigen. CAR-TIL produced greater amounts of IFN compared with parental TIL, when co-cultured with Her2 expressing tumor lines, including autologous melanoma tumor lines, although no consistent increase in cytotoxicity by TIL was afforded by expression of a CAR. Results of an in vivo study in NSG mice demonstrated tumor shrinkage when CAR-TILs were used in an adoptive cell therapy protocol. Conclusion: Potential limitations of transduced TIL in our study included limited proliferative potential and a terminally differentiated phenotype, which would need addressing in further work before consideration of clinical translation.

19.
Clin Cancer Res ; 27(22): 6222-6234, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34475103

RESUMEN

PURPOSE: In this article, we describe a combination chimeric antigen receptor (CAR) T-cell therapy that eradicated the majority of tumors in two immunocompetent murine pancreatic cancer models and a human pancreatic cancer xenograft model. EXPERIMENTAL DESIGN: We used a dual-specific murine CAR T cell that expresses a CAR against the Her2 tumor antigen, and a T-cell receptor (TCR) specific for gp100. As gp100 is also known as pMEL, the dual-specific CAR T cells are thus denoted as CARaMEL cells. A vaccine containing live vaccinia virus coding a gp100 minigene (VV-gp100) was administered to the recipient mice to stimulate CARaMEL cells. The treatment also included the histone deacetylase inhibitor panobinostat (Pano). RESULTS: The combination treatment enabled significant suppression of Her2+ pancreatic cancers leading to the eradication of the majority of the tumors. Besides inducing cancer cell apoptosis, Pano enhanced CAR T-cell gene accessibility and promoted CAR T-cell differentiation into central memory cells. To test the translational potential of this approach, we established a method to transduce human T cells with an anti-Her2 CAR and a gp100-TCR. The exposure of the human T cells to Pano promoted a T-cell central memory phenotype and the combination treatment of human CARaMEL cells and Pano eradicated human pancreatic cancer xenografts in mice. CONCLUSIONS: We propose that patients with pancreatic cancer could be treated using a scheme that contains dual-specific CAR T cells, a vaccine that activates the dual-specific CAR T cells through their TCR, and the administration of Pano.


Asunto(s)
Neoplasias Pancreáticas , Receptores Quiméricos de Antígenos , Animales , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Inmunoterapia Adoptiva/métodos , Ratones , Neoplasias Pancreáticas/terapia , Panobinostat , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Cancer Immunol Immunother ; 59(8): 1235-46, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20376439

RESUMEN

Natural killer (NK) cells represent a promising cell type to utilize for effective adoptive immunotherapy. However, little is known about the important cytolytic molecules and signaling pathways used by NK cells in the adoptive transfer setting. To address this issue, we developed a novel mouse model to investigate the trafficking and mechanism of action of these cells. We demonstrate that methylcholanthrene-induced RKIK sarcoma cells were susceptible to NK cell-mediated lysis in vitro and in vivo following adoptive transfer of NK cells in C57BL/6 RAG-2(-/-)gammac(-/-) mice. Cytotoxic molecules perforin, granzymes B and M as well as the death ligand TRAIL and pro-inflammatory cytokine IFN-gamma were found to be important in the anti-tumor effect mediated by adoptively transferred NK cells. Importantly, we demonstrate that adoptively transferred NK cells could traffic to the tumor site and persisted in vivo which correlated with the anti-tumor effect observed. Overall, the results of this study have important implications for enhancing NK cell-based immunotherapies.


Asunto(s)
Citotoxicidad Inmunológica , Inmunoterapia Adoptiva , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Sarcoma Experimental/inmunología , Sarcoma Experimental/terapia , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Interleucina-2/inmunología , Interleucina-2/metabolismo , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Metilcolantreno , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Sarcoma Experimental/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA