Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Immunol ; 202(4): 1301-1310, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30642983

RESUMO

Characterizing self-tolerance mechanisms and their failure is critical to understand immune homeostasis, cancer immunity, and autoimmunity. However, examination of self-tolerance mechanisms has relied primarily on transgenic mice expressing TCRs targeting well-characterized, but nonphysiologic, model Ags, such as OVA and hemagglutinin. Identifying TCRs directed against bona fide self-antigens is made difficult by the extraordinary diversity of TCRs and the low prevalence of Ag-specific clones (<10-100 naive cells per organism), limiting dissection of tolerance mechanisms restricting immunity to self-proteins. In this study, we isolated and characterized TCRs recognizing the intestinal epithelial cell receptor and colorectal cancer Ag GUCY2C to establish a model to study self-antigen-specific tolerance mechanisms. GUCY2C-specific CD4+ effector T cells were isolated from immunized, nontolerant Gucy2c -/- mice. Next-generation sequencing identified GUCY2C-specific TCRs, which were engineered into CD4+ T cells in vitro to confirm TCR recognition of GUCY2C. Further, the generation of "retrogenic" mice by reconstitution with TCR-transduced hematopoietic stem cells resulted in normal CD4+ T cell development, responsiveness to immunization, and GUCY2C-induced tolerance in recipient mice, recapitulating observations in conventional models. This retrogenic model can be employed to define self-tolerance mechanisms restricting T and B cell responses to GUCY2C to optimize colorectal cancer immunotherapy without autoimmunity.


Assuntos
Neoplasias Colorretais/imunologia , Modelos Imunológicos , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Enterotoxina/imunologia , Animais , Feminino , Tolerância Imunológica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos
2.
Nature ; 522(7556): 315-20, 2015 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-26085270

RESUMO

There is an urgent need for new drugs to treat malaria, with broad therapeutic potential and novel modes of action, to widen the scope of treatment and to overcome emerging drug resistance. Here we describe the discovery of DDD107498, a compound with a potent and novel spectrum of antimalarial activity against multiple life-cycle stages of the Plasmodium parasite, with good pharmacokinetic properties and an acceptable safety profile. DDD107498 demonstrates potential to address a variety of clinical needs, including single-dose treatment, transmission blocking and chemoprotection. DDD107498 was developed from a screening programme against blood-stage malaria parasites; its molecular target has been identified as translation elongation factor 2 (eEF2), which is responsible for the GTP-dependent translocation of the ribosome along messenger RNA, and is essential for protein synthesis. This discovery of eEF2 as a viable antimalarial drug target opens up new possibilities for drug discovery.


Assuntos
Antimaláricos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Malária/parasitologia , Plasmodium/efeitos dos fármacos , Plasmodium/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Quinolinas/farmacologia , Animais , Antimaláricos/administração & dosagem , Antimaláricos/efeitos adversos , Antimaláricos/farmacocinética , Descoberta de Drogas , Feminino , Estágios do Ciclo de Vida/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/parasitologia , Malária/tratamento farmacológico , Masculino , Modelos Moleculares , Fator 2 de Elongação de Peptídeos/antagonistas & inibidores , Fator 2 de Elongação de Peptídeos/metabolismo , Plasmodium/genética , Plasmodium/crescimento & desenvolvimento , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/fisiologia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Plasmodium vivax/efeitos dos fármacos , Plasmodium vivax/metabolismo , Quinolinas/administração & dosagem , Quinolinas/química , Quinolinas/farmacocinética
3.
J Virol ; 91(2)2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27807241

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) emerged in 2012 and is a highly pathogenic respiratory virus. There are no treatment options against MERS-CoV for humans or animals, and there are no large-scale clinical trials for therapies against MERS-CoV. To address this need, we developed an inactivated rabies virus (RABV) that contains the MERS-CoV spike (S) protein expressed on its surface. Our initial recombinant vaccine, BNSP333-S, expresses a full-length wild-type MERS-CoV S protein; however, it showed significantly reduced viral titers compared to those of the parental RABV strain and only low-level incorporation of full-length MERS-CoV S into RABV particles. Therefore, we developed a RABV-MERS vector that contained the MERS-CoV S1 domain of the MERS-CoV S protein fused to the RABV G protein C terminus (BNSP333-S1). BNSP333-S1 grew to titers similar to those of the parental vaccine vector BNSP333, and the RABV G-MERS-CoV S1 fusion protein was efficiently expressed and incorporated into RABV particles. When we vaccinated mice, chemically inactivated BNSP333-S1 induced high-titer neutralizing antibodies. Next, we challenged both vaccinated mice and control mice with MERS-CoV after adenovirus transduction of the human dipeptidyl peptidase 4 (hDPP4) receptor and then analyzed the ability of mice to control MERS-CoV infection. Our results demonstrated that vaccinated mice were fully protected from the MERS-CoV challenge, as indicated by the significantly lower MERS-CoV titers and MERS-CoV and mRNA levels in challenged mice than those in unvaccinated controls. These data establish that an inactivated RABV-MERS S-based vaccine may be effective for use in animals and humans in areas where MERS-CoV is endemic. IMPORTANCE: Rabies virus-based vectors have been proven to be efficient dual vaccines against rabies and emergent infectious diseases such as Ebola virus. Here we show that inactivated rabies virus particles containing the MERS-CoV S1 protein induce potent immune responses against MERS-CoV and RABV. This novel vaccine is easy to produce and may be useful to protect target animals, such as camels, as well as humans from deadly MERS-CoV and RABV infections. Our results indicate that this vaccine approach can prevent disease, and the RABV-based vaccine platform may be a valuable tool for timely vaccine development against emerging infectious diseases.


Assuntos
Infecções por Coronavirus/imunologia , Proteção Cruzada/imunologia , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Vacinas Virais/imunologia , Animais , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Modelos Animais de Doenças , Regulação Viral da Expressão Gênica , Humanos , Imunização , Camundongos , Interações Microbianas , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Raiva/prevenção & controle , Raiva/virologia , Vírus da Raiva/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinas Atenuadas , Vacinas Sintéticas , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Vacinas Virais/genética , Montagem de Vírus
5.
J Immunother Cancer ; 7(1): 104, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31010434

RESUMO

BACKGROUND: The colorectal cancer antigen GUCY2C exhibits unique split tolerance, evoking antigen-specific CD8+, but not CD4+, T-cell responses that deliver anti-tumor immunity without autoimmunity in mice. Here, the cancer vaccine Ad5-GUCY2C-PADRE was evaluated in a first-in-man phase I clinical study of patients with early-stage colorectal cancer to assess its safety and immunological efficacy. METHODS: Ten patients with surgically-resected stage I or stage II (pN0) colon cancer received a single intramuscular injection of 1011 viral particles (vp) of Ad5-GUCY2C-PADRE. Safety assessment and immunomonitoring were carried out for 6 months following immunization. This trial employed continual monitoring of both efficacy and toxicity of subjects as joint primary outcomes. RESULTS: All patients receiving Ad5-GUCY2C-PADRE completed the study and none developed adverse events greater than grade 1. Antibody responses to GUCY2C were detected in 10% of patients, while 40% exhibited GUCY2C-specific T-cell responses. GUCY2C-specific responses were exclusively CD8+ cytotoxic T cells, mimicking pre-clinical studies in mice in which GUCY2C-specific CD4+ T cells are eliminated by self-tolerance, while CD8+ T cells escape tolerance and mediate antitumor immunity. Moreover, pre-existing neutralizing antibodies (NAbs) to the Ad5 vector were associated with poor vaccine-induced responses, suggesting that Ad5 NAbs oppose GUCY2C immune responses to the vaccine in patients and supported by mouse studies. CONCLUSIONS: Split tolerance to GUCY2C in cancer patients can be exploited to safely generate antigen-specific cytotoxic CD8+, but not autoimmune CD4+, T cells by Ad5-GUCY2C-PADRE in the absence of pre-existing NAbs to the viral vector. TRIAL REGISTRATION: This trial (NCT01972737) was registered at ClinicalTrials.gov on October 30th, 2013. https://clinicaltrials.gov/ct2/show/NCT01972737.


Assuntos
Vacinas Anticâncer/efeitos adversos , Neoplasias Colorretais/terapia , Imunoterapia/métodos , Receptores de Enterotoxina/imunologia , Linfócitos T Citotóxicos/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Idoso , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Linfócitos T CD4-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/imunologia , Colo/patologia , Colo/cirurgia , Neoplasias Colorretais/sangue , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Terapia Combinada/métodos , Relação Dose-Resposta Imunológica , Feminino , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Humanos , Tolerância Imunológica , Imunogenicidade da Vacina , Masculino , Camundongos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Receptores de Enterotoxina/genética , Reto/patologia , Reto/cirurgia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
6.
Cancer Immunol Res ; 6(5): 509-516, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29615399

RESUMO

One major hurdle to the success of adoptive T-cell therapy is the identification of antigens that permit effective targeting of tumors in the absence of toxicities to essential organs. Previous work has demonstrated that T cells engineered to express chimeric antigen receptors (CAR-T cells) targeting the murine homolog of the colorectal cancer antigen GUCY2C treat established colorectal cancer metastases, without toxicity to the normal GUCY2C-expressing intestinal epithelium, reflecting structural compartmentalization of endogenous GUCY2C to apical membranes comprising the intestinal lumen. Here, we examined the utility of a human-specific, GUCY2C-directed single-chain variable fragment as the basis for a CAR construct targeting human GUCY2C-expressing metastases. Human GUCY2C-targeted murine CAR-T cells promoted antigen-dependent T-cell activation quantified by activation marker upregulation, cytokine production, and killing of GUCY2C-expressing, but not GUCY2C-deficient, cancer cells in vitro GUCY2C CAR-T cells provided long-term protection against lung metastases of murine colorectal cancer cells engineered to express human GUCY2C in a syngeneic mouse model. GUCY2C murine CAR-T cells recognized and killed human colorectal cancer cells endogenously expressing GUCY2C, providing durable survival in a human xenograft model in immunodeficient mice. Thus, we have identified a human GUCY2C-specific CAR-T cell therapy approach that may be developed for the treatment of GUCY2C-expressing metastatic colorectal cancer. Cancer Immunol Res; 6(5); 509-16. ©2018 AACR.


Assuntos
Neoplasias Colorretais/terapia , Citotoxicidade Imunológica , Imunoterapia Adotiva/métodos , Neoplasias Pulmonares/prevenção & controle , Receptores de Antígenos Quiméricos/metabolismo , Receptores de Enterotoxina , Linfócitos T/transplante , Animais , Células Cultivadas , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Metástase Neoplásica , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/genética , Receptores de Enterotoxina/genética , Receptores de Enterotoxina/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Per Med ; 14(3): 259-270, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-29767586

RESUMO

Cancer immunotherapy has long offered the promise of producing cancer treatments that are more effective and less toxic than traditional chemotherapy and radiotherapy. That potential has only begun to be realized in the last 5 years with the first US FDA-approved cancer vaccine (sipuleucel-T), checkpoint inhibitors and adoptive cell therapy. While these therapies have been remarkably more effective than previous cancer immunotherapeutics, they are often limited by their inherently personalized nature. Indeed, each patient's immune system and cancer are unique, limiting the scalability and generalizability of new approaches. However, emerging solutions may overcome these limitations, producing 'off-the-shelf' cancer immunotherapies that transform patient outcomes.


Assuntos
Imunoterapia Adotiva/estatística & dados numéricos , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Vacinas Anticâncer/imunologia , Humanos , Imunoterapia Adotiva/métodos , Medicina de Precisão/tendências
8.
Sci Transl Med ; 9(387)2017 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-28446690

RESUMO

As part of the global effort toward malaria eradication, phenotypic whole-cell screening revealed the 2-aminopyridine class of small molecules as a good starting point to develop new antimalarial drugs. Stemming from this series, we found that the derivative, MMV390048, lacked cross-resistance with current drugs used to treat malaria. This compound was efficacious against all Plasmodium life cycle stages, apart from late hypnozoites in the liver. Efficacy was shown in the humanized Plasmodium falciparum mouse model, and modest reductions in mouse-to-mouse transmission were achieved in the Plasmodium berghei mouse model. Experiments in monkeys revealed the ability of MMV390048 to be used for full chemoprotection. Although MMV390048 was not able to eliminate liver hypnozoites, it delayed relapse in a Plasmodium cynomolgi monkey model. Both genomic and chemoproteomic studies identified a kinase of the Plasmodium parasite, phosphatidylinositol 4-kinase, as the molecular target of MMV390048. The ability of MMV390048 to block all life cycle stages of the malaria parasite suggests that this compound should be further developed and may contribute to malaria control and eradication as part of a single-dose combination treatment.


Assuntos
1-Fosfatidilinositol 4-Quinase/antagonistas & inibidores , Aminopiridinas/uso terapêutico , Antimaláricos/uso terapêutico , Sulfonas/uso terapêutico , Aminopiridinas/farmacologia , Animais , Antimaláricos/farmacologia , Feminino , Malária/tratamento farmacológico , Malária/enzimologia , Masculino , Camundongos , Camundongos SCID , Testes de Sensibilidade Parasitária , Plasmodium/efeitos dos fármacos , Plasmodium/patogenicidade , Sulfonas/farmacologia
9.
Oncoimmunology ; 5(10): e1227897, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27853651

RESUMO

Adoptive T-cell therapy (ACT) is an emerging paradigm in which T cells are genetically modified to target cancer-associated antigens and eradicate tumors. However, challenges treating epithelial cancers with ACT reflect antigen targets that are not tumor-specific, permitting immune damage to normal tissues, and preclinical testing in artificial xenogeneic models, preventing prediction of toxicities in patients. In that context, mucosa-restricted antigens expressed by cancers exploit anatomical compartmentalization which shields mucosae from systemic antitumor immunity. This shielding may be amplified with ACT platforms employing antibody-based chimeric antigen receptors (CARs), which mediate MHC-independent recog-nition of antigens. GUCY2C is a cancer mucosa antigen expressed on the luminal surfaces of the intestinal mucosa in mice and humans, and universally overexpressed by colorectal tumors, suggesting its unique utility as an ACT target. T cells expressing CARs directed by a GUCY2C-specific antibody fragment recognized GUCY2C, quantified by expression of activation markers and cytokines. Further, GUCY2C CAR-T cells lysed GUCY2C-expressing, but not GUCY2C-deficient, mouse colorectal cancer cells. Moreover, GUCY2C CAR-T cells reduced tumor number and morbidity and improved survival in mice harboring GUCY2C-expressing colorectal cancer metastases. GUCY2C-directed T cell efficacy reflected CAR affinity and surface expression and was achieved without immune-mediated damage to normal tissues in syngeneic mice. These observations highlight the potential for therapeutic translation of GUCY2C-directed CAR-T cells to treat metastatic tumors, without collateral autoimmunity, in patients with metastatic colorectal cancer.

10.
Arch Dermatol ; 147(12): 1403-9, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22184762

RESUMO

OBJECTIVE: To report the financial impact of diagnosing and treating the dermatologic toxicities (dTs) that develop in patients receiving targeted anticancer therapies. DESIGN: Single-center retrospective and prospective medical record data extraction. SETTING: Department of Dermatology, Northwestern University, Chicago, Illinois. PATIENTS: One hundred thirty-two adults who presented between November 1, 2005, and June 30, 2008, and who were diagnosed as having 1 primary cancer type and were treated with 1 molecularly targeted agent. MAIN OUTCOME MEASURE: Standard billable costs to the patient for dT-related medications, clinic visits, laboratory and diagnostic testing, and therapeutic procedures. RESULTS: The 132 patients had a median of 3 clinic visits for dT management with a median cost of $1920 per patient. Sorafenib was associated with the most costly overall median cost per patient ($2509 per patient), and imatinib was associated with the least costly overall median cost per patient ($1263 per patient). Among the 7 targeted drugs and all 10 dTs, the most costly dT (measured by cost of treatment with medications) was hand/foot skin reaction, associated with sorafenib therapy (median cost, $968 per patient) (P < .001). The second most costly dT was panitumumab-associated acneiform eruption (median cost, $933 per patient) (P < .001). CONCLUSION: The cost of diagnosis and treatment of dTs associated with targeted agents contributes to the overall economic burden of cancer care. Efforts toward the prevention of dTs may be important for decreasing the financial burden in oncology.


Assuntos
Antineoplásicos/efeitos adversos , Toxidermias/economia , Toxidermias/etiologia , Custos de Cuidados de Saúde , Terapia de Alvo Molecular/efeitos adversos , Neoplasias/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Assistência Ambulatorial/economia , Assistência Ambulatorial/estatística & dados numéricos , Antineoplásicos/economia , Antineoplásicos/uso terapêutico , Custos de Medicamentos , Toxidermias/diagnóstico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular/economia , Terapia de Alvo Molecular/métodos , Neoplasias/patologia , Estudos Prospectivos , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA