Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
MAbs ; 13(1): 1850395, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33459147

RESUMEN

We report here the discovery and optimization of a novel T cell retargeting anti-GUCY2C x anti-CD3ε bispecific antibody for the treatment of solid tumors. Using a combination of hybridoma, phage display and rational design protein engineering, we have developed a fully humanized and manufacturable CD3 bispecific antibody that demonstrates favorable pharmacokinetic properties and potent in vivo efficacy. Anti-GUCY2C and anti-CD3ε antibodies derived from mouse hybridomas were first humanized into well-behaved human variable region frameworks with full retention of binding and T-cell mediated cytotoxic activity. To address potential manufacturability concerns, multiple approaches were taken in parallel to optimize and de-risk the two antibody variable regions. These approaches included structure-guided rational mutagenesis and phage display-based optimization, focusing on improving stability, reducing polyreactivity and self-association potential, removing chemical liabilities and proteolytic cleavage sites, and de-risking immunogenicity. Employing rapid library construction methods as well as automated phage display and high-throughput protein production workflows enabled efficient generation of an optimized bispecific antibody with desirable manufacturability properties, high stability, and low nonspecific binding. Proteolytic cleavage and deamidation in complementarity-determining regions were also successfully addressed. Collectively, these improvements translated to a molecule with potent single-agent in vivo efficacy in a tumor cell line adoptive transfer model and a cynomolgus monkey pharmacokinetic profile (half-life>4.5 days) suitable for clinical development. Clinical evaluation of PF-07062119 is ongoing.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Complejo CD3/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores de Enterotoxina/inmunología , Animales , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Biespecíficos/uso terapéutico , Línea Celular Tumoral , Femenino , Humanos , Hibridomas , Macaca fascicularis/inmunología , Macaca fascicularis/metabolismo , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/metabolismo , Ingeniería de Proteínas/métodos , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/farmacocinética , Anticuerpos de Cadena Única/uso terapéutico , Linfocitos T/inmunología , Linfocitos T/metabolismo
2.
Clin Cancer Res ; 26(9): 2188-2202, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31996389

RESUMEN

PURPOSE: Gastrointestinal cancers remain areas of high unmet need despite advances in targeted and immunotherapies. Here, we demonstrate potent, tumor-selective efficacy with PF-07062119, a T-cell engaging CD3 bispecific targeting tumors expressing Guanylyl Cyclase C (GUCY2C), which is expressed widely across colorectal cancer and other gastrointestinal malignancies. In addition, to address immune evasion mechanisms, we explore combinations with immune checkpoint blockade agents and with antiangiogenesis therapy. EXPERIMENTAL DESIGN: PF-07062119 activity was evaluated in vitro in multiple tumor cell lines, and in vivo in established subcutaneous and orthotopic human colorectal cancer xenograft tumors with adoptive transfer of human T cells. Efficacy was also evaluated in mouse syngeneic tumors using human CD3ε transgenic mice. IHC and mass cytometry were performed to demonstrate drug biodistribution, recruitment of activated T cells, and to identify markers of immune evasion. Combination studies were performed with anti-PD-1/PD-L1 and anti-VEGF antibodies. Toxicity and pharmacokinetic studies were done in cynomolgus macaque. RESULTS: We demonstrate that GUCY2C-positive tumors can be targeted with an anti-GUCY2C/anti-CD3ε bispecific, with selective drug biodistribution to tumors. PF-07062119 showed potent T-cell-mediated in vitro activity and in vivo efficacy in multiple colorectal cancer human xenograft tumor models, including KRAS- and BRAF-mutant tumors, as well as in the immunocompetent mouse syngeneic tumor model. PF-07062119 activity was further enhanced when combined with anti-PD-1/PD-L1 treatment or in combination with antiangiogenic therapy. Toxicity studies in cynomolgus indicated a monitorable and manageable toxicity profile. CONCLUSIONS: These data highlight the potential for PF-07062119 to demonstrate efficacy and improve patient outcomes in colorectal cancer and other gastrointestinal malignancies.


Asunto(s)
Anticuerpos Biespecíficos/administración & dosificación , Complejo CD3/inmunología , Neoplasias Colorrectales/terapia , Neoplasias Gastrointestinales/terapia , Inmunoterapia/métodos , Receptores de Enterotoxina/inmunología , Linfocitos T/inmunología , Traslado Adoptivo/métodos , Animales , Anticuerpos Biespecíficos/farmacocinética , Línea Celular Tumoral , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/metabolismo , Modelos Animales de Enfermedad , Femenino , Neoplasias Gastrointestinales/inmunología , Neoplasias Gastrointestinales/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Distribución Tisular
3.
J Immunother Cancer ; 7(1): 104, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31010434

RESUMEN

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.


Asunto(s)
Vacunas contra el Cáncer/efectos adversos , Neoplasias Colorrectales/terapia , Inmunoterapia/métodos , Receptores de Enterotoxina/inmunología , Linfocitos T Citotóxicos/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Anciano , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Colon/patología , Colon/cirugía , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Terapia Combinada/métodos , Relación Dosis-Respuesta Inmunológica , Femenino , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Tolerancia Inmunológica , Inmunogenicidad Vacunal , Masculino , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Receptores de Enterotoxina/genética , Recto/patología , Recto/cirugía , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/inmunología
4.
Psychopharmacology (Berl) ; 236(10): 2997-3011, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30806743

RESUMEN

Depression is a common comorbidity in cancer cases, but this is not only due to the emotional distress of having a life-threatening disease. A common biological mechanism, involving a dysregulated immune system, seems to underpin this comorbidity. In particular, the activation of the kynurenine pathway of tryptophan degradation due to inflammation may play a key role in the development and persistence of both diseases. As a consequence, targeting enzymes involved in this pathway offers a unique opportunity to develop new strategies to treat cancer and depression at once. In this work, we provide a systematic review of the evidence up to date on the kynurenine pathway role in linking depression and cancer and on clinical implications of this evidence. In particular, complications due to chemotherapy are discussed, as well as the potential antidepressant efficacy of novel immunotherapies for cancer.


Asunto(s)
Depresión/metabolismo , Quinurenina/metabolismo , Neoplasias/metabolismo , Transducción de Señal/fisiología , Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Depresión/inmunología , Humanos , Inmunoterapia/métodos , Inmunoterapia/tendencias , Indolamina-Pirrol 2,3,-Dioxigenasa/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/uso terapéutico , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Quinurenina/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Receptores de Enterotoxina/inmunología , Receptores de Enterotoxina/metabolismo , Transducción de Señal/efectos de los fármacos
5.
J Immunol ; 202(4): 1301-1310, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30642983

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/inmunología , Modelos Inmunológicos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Enterotoxina/inmunología , Animales , Femenino , Tolerancia Inmunológica/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos
6.
Cancer Immunol Res ; 6(5): 509-516, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29615399

RESUMEN

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.


Asunto(s)
Neoplasias Colorrectales/terapia , Citotoxicidad Inmunológica , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/prevención & control , Receptores Quiméricos de Antígenos/metabolismo , Receptores de Enterotoxina , Linfocitos T/trasplante , Animales , Células Cultivadas , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Metástasis de la Neoplasia , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores de Enterotoxina/genética , Receptores de Enterotoxina/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS One ; 13(1): e0191046, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29370189

RESUMEN

Guanylyl cyclase C (GCC) is a cell-surface protein that is expressed by normal intestinal epithelial cells, more than 95% of metastatic colorectal cancers (mCRC), and the majority of gastric and pancreatic cancers. Due to strict apical localization, systemically delivered GCC-targeting agents should not reach GCC in normal intestinal tissue, while accessing antigen in tumor. We generated an investigational antibody-drug conjugate (TAK-264, formerly MLN0264) comprising a fully human anti-GCC monoclonal antibody conjugated to monomethyl auristatin E via a protease-cleavable peptide linker. TAK-264 specifically bound, was internalized by, and killed GCC-expressing cells in vitro in an antigen-density-dependent manner. In GCC-expressing xenograft models with similar GCC expression levels/patterns observed in human mCRC samples, TAK-264 induced cell death, leading to tumor regressions and long-term tumor growth inhibition. TAK-264 antitumor activity was generally antigen-density-dependent, although some GCC-expressing tumors were refractory to TAK-264-targeted high local concentrations of payload. These data support further evaluation of TAK-264 in the treatment of GCC-expressing tumors.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunoconjugados/farmacología , Oligopéptidos/metabolismo , Receptores de Enterotoxina/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados , Western Blotting , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Femenino , Células HEK293 , Humanos , Mucosa Intestinal/enzimología , Ratones , Ratones SCID , Receptores de Enterotoxina/genética , Receptores de Enterotoxina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Invest New Drugs ; 35(2): 235-241, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28188407

RESUMEN

Background The first-in-class antibody-drug conjugate TAK-264 (formerly MLN0264) consists of an antibody targeting guanylyl cyclase C (GCC) conjugated to monomethyl auristatin E (MMAE) via a peptide linker. This phase II study evaluated the efficacy and safety of TAK-264 in patients with adenocarcinoma of the stomach or gastroesophageal junction expressing GCC, who had progressed on ≥1 line of prior therapy. Methods This study used a two-stage design, with an interim analysis conducted after stage I to determine whether to continue to stage II or discontinue on the grounds of futility. Adult patients with gastric and gastroesophageal junction adenocarcinoma expressing low, intermediate, or high GCC levels received TAK-264 1.8 mg/kg as a 30-min intravenous infusion once every 21 days, for up to 1 year. The primary endpoint was objective response rate. Radiographic assessments of tumor burden were performed every 2 cycles (6 weeks). Results A total of 38 patients participated in the study. Patients received a median of 2 (range 1-14) cycles; 8 (21%) received at least 6 cycles. The most common adverse events were nausea (53%), fatigue (32%), and decreased appetite (29%). Grade ≥3 events including anemia, diarrhea, and neutropenia were seen in 14 (37%) patients. Systemic exposure to TAK-264 was maintained throughout each treatment cycle. Two patients (6%) with intermediate GCC expression had objective responses. Conclusions TAK-264 demonstrated a manageable safety profile in this patient population. The stage I interim analysis did not support continuation to stage II of the study.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Neoplasias Esofágicas/tratamiento farmacológico , Inmunoconjugados/uso terapéutico , Receptores de Enterotoxina/inmunología , Neoplasias Gástricas/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Unión Esofagogástrica/patología , Femenino , Humanos , Inmunoconjugados/efectos adversos , Inmunoconjugados/farmacocinética , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Receptores de Enterotoxina/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...