Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 1900, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38429261

ABSTRACT

Although co-stimulation of T cells with agonist antibodies targeting 4-1BB (CD137) improves antitumor immune responses in preclinical studies, clinical success has been limited by on-target, off-tumor activity. Here, we report the development of a tumor-anchored ɑ4-1BB agonist (ɑ4-1BB-LAIR), which consists of a ɑ4-1BB antibody fused to the collagen-binding protein LAIR. While combination treatment with an antitumor antibody (TA99) shows only modest efficacy, simultaneous depletion of CD4+ T cells boosts cure rates to over 90% of mice. Mechanistically, this synergy depends on ɑCD4 eliminating tumor draining lymph node regulatory T cells, resulting in priming and activation of CD8+ T cells which then infiltrate the tumor microenvironment. The cytotoxic program of these newly primed CD8+ T cells is then supported by the combined effect of TA99 and ɑ4-1BB-LAIR. The combination of TA99 and ɑ4-1BB-LAIR with a clinically approved ɑCTLA-4 antibody known for enhancing T cell priming results in equivalent cure rates, which validates the mechanistic principle, while the addition of ɑCTLA-4 also generates robust immunological memory against secondary tumor rechallenge. Thus, our study establishes the proof of principle for a clinically translatable cancer immunotherapy.


Subject(s)
Antineoplastic Agents , Neoplasms , T-Lymphocytes, Regulatory , Animals , Mice , Antibodies , CD8-Positive T-Lymphocytes , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Tumor Microenvironment , 4-1BB Ligand/immunology
2.
Commun Med (Lond) ; 3(1): 108, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37558833

ABSTRACT

BACKGROUND: Genetically engineered mouse models (GEMMs) of cancer are powerful tools to study mechanisms of disease progression and therapy response, yet little is known about how these models respond to multimodality therapy used in patients. Radiation therapy (RT) is frequently used to treat localized cancers with curative intent, delay progression of oligometastases, and palliate symptoms of metastatic disease. METHODS: Here we report the development, testing, and validation of a platform to immobilize and target tumors in mice with stereotactic ablative RT (SART). Xenograft and autochthonous tumor models were treated with hypofractionated ablative doses of radiotherapy. RESULTS: We demonstrate that hypofractionated regimens used in clinical practice can be effectively delivered in mouse models. SART alters tumor stroma and the immune environment, improves survival in GEMMs of primary prostate and colorectal cancer, and synergizes with androgen deprivation in prostate cancer. Complete pathologic responses were achieved in xenograft models, but not in GEMMs. CONCLUSIONS: While SART is capable of fully ablating xenografts, it is unable to completely eradicate disease in GEMMs, arguing that resistance to potentially curative therapy can be modeled in GEMMs.


Mice can be used to model the types of cancer seen in people to investigate the effects of cancer therapies, such as radiation. Here, we apply radiation therapy treatments that are able to cure cancer in humans to mice that have cancer of the prostate or colorectum. We show that the mice do not experience many side effects and that the tumours reduce in size, but in some cases show progression after treatment. Our study demonstrates that mice can be used to better understand how human cancers respond to radiation treatment, which can lead to the development of improved treatments and treatment schedules.

3.
Sci Rep ; 5: 14759, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26440059

ABSTRACT

Personalized cancer medicine requires measurement of therapeutic efficacy as early as possible, which is optimally achieved by three-dimensional imaging given the heterogeneity of cancer. Magnetic resonance imaging (MRI) can obtain images of both anatomy and cellular responses, if acquired with a molecular imaging contrast agent. The poor sensitivity of MRI has limited the development of activatable molecular MR contrast agents. To overcome this limitation of molecular MRI, a novel implementation of our caspase-3-sensitive nanoaggregation MRI (C-SNAM) contrast agent is reported. C-SNAM is triggered to self-assemble into nanoparticles in apoptotic tumor cells, and effectively amplifies molecular level changes through nanoaggregation, enhancing tissue retention and spin-lattice relaxivity. At one-tenth the current clinical dose of contrast agent, and following a single imaging session, C-SNAM MRI accurately measured the response of tumors to either metronomic chemotherapy or radiation therapy, where the degree of signal enhancement is prognostic of long-term therapeutic efficacy. Importantly, C-SNAM is inert to immune activation, permitting radiation therapy monitoring.


Subject(s)
Contrast Media , Magnetic Resonance Imaging/methods , Neoplasms/drug therapy , Neoplasms/pathology , Administration, Metronomic , Animals , Antibiotics, Antineoplastic/administration & dosage , Caspase 3/metabolism , Contrast Media/chemistry , Cysteine/administration & dosage , Cysteine/analogs & derivatives , Cysteine/chemistry , Doxorubicin/administration & dosage , Female , HeLa Cells , Humans , Mice, Nude , Neoplasms/radiotherapy , Quinolines/administration & dosage , Quinolines/chemistry , Treatment Outcome , Xenograft Model Antitumor Assays/methods
5.
J Theor Biol ; 314: 57-68, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-22974563

ABSTRACT

Antibodies are under development to treat a variety of cancers, such as lymphomas, colon, and breast cancer. A major limitation to greater efficacy for this class of drugs is poor distribution in vivo. Localization of antibodies occurs slowly, often in insufficient therapeutic amounts, and distributes heterogeneously throughout the tumor. While the microdistribution around individual vessels is important for many therapies, the total amount of antibody localized in the tumor is paramount for many applications such as imaging, determining the therapeutic index with antibody drug conjugates, and dosing in radioimmunotherapy. With imaging and pretargeted therapeutic strategies, the time course of uptake is critical in determining when to take an image or deliver a secondary reagent. We present here a simple mechanistic model of antibody uptake and retention that captures the major rates that determine the time course of antibody concentration within a tumor including dose, affinity, plasma clearance, target expression, internalization, permeability, and vascularization. Since many of the parameters are known or can be estimated in vitro, this model can approximate the time course of antibody concentration in tumors to aid in experimental design, data interpretation, and strategies to improve localization.


Subject(s)
Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/metabolism , Cell Compartmentation , Models, Biological , Neoplasms/immunology , Neoplasms/metabolism , Animals , Antibodies, Neoplasm/blood , Antibodies, Neoplasm/therapeutic use , Computer Simulation , Humans , Immunoglobulin G/immunology , Mice , Neoplasms/blood , Neoplasms/drug therapy , Reproducibility of Results , Single-Chain Antibodies/immunology , Time Factors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL