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










Publication year range
1.
Blood Adv ; 8(7): 1747-1759, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38324726

ABSTRACT

ABSTRACT: Therapeutic vaccination has long been a promising avenue for cancer immunotherapy but is often limited by tumor heterogeneity. The genetic and molecular diversity between patients often results in variation in the antigens present on cancer cell surfaces. As a result, recent research has focused on personalized cancer vaccines. Although promising, this strategy suffers from time-consuming production, high cost, inaccessibility, and targeting of a limited number of tumor antigens. Instead, we explore an antigen-agnostic polymeric in situ cancer vaccination platform for treating blood malignancies, in our model here with acute myeloid leukemia (AML). Rather than immunizing against specific antigens or targeting adjuvant to specific cell-surface markers, this platform leverages a characteristic metabolic and enzymatic dysregulation in cancer cells that produces an excess of free cysteine thiols on their surfaces. These thiols increase in abundance after treatment with cytotoxic agents such as cytarabine, the current standard of care in AML. The resulting free thiols can undergo efficient disulfide exchange with pyridyl disulfide (PDS) moieties on our construct and allow for in situ covalent attachment to cancer cell surfaces and debris. PDS-functionalized monomers are incorporated into a statistical copolymer with pendant mannose groups and TLR7 agonists to target covalently linked antigen and adjuvant to antigen-presenting cells in the liver and spleen after IV administration. There, the compound initiates an anticancer immune response, including T-cell activation and antibody generation, ultimately prolonging survival in cancer-bearing mice.


Subject(s)
Cysteine , Leukemia, Myeloid, Acute , Humans , Mice , Animals , Cysteine/therapeutic use , Disease Models, Animal , Leukemia, Myeloid, Acute/drug therapy , Adjuvants, Immunologic , Antigens, Neoplasm , Lymphocyte Activation , Disulfides/therapeutic use
2.
Chem Soc Rev ; 53(4): 1789-1822, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38170619

ABSTRACT

Immunoengineering is a rapidly evolving field that has been driving innovations in manipulating immune system for new treatment tools and methods. The need for materials for immunoengineering applications has gained significant attention in recent years due to the growing demand for effective therapies that can target and regulate the immune system. Biologics and biomaterials are emerging as promising tools for controlling immune responses, and a wide variety of materials, including proteins, polymers, nanoparticles, and hydrogels, are being developed for this purpose. In this review article, we explore the different types of materials used in immunoengineering applications, their properties and design principles, and highlight the latest therapeutic materials advancements. Recent works in adjuvants, vaccines, immune tolerance, immunotherapy, and tissue models for immunoengineering studies are discussed.


Subject(s)
Immunotherapy , Vaccines , Biocompatible Materials/therapeutic use , Proteins
3.
Sci Adv ; 9(48): eadh9879, 2023 12.
Article in English | MEDLINE | ID: mdl-38019919

ABSTRACT

Cancer immunotherapy is moving toward combination regimens with agents of complementary mechanisms of action to achieve more frequent and robust efficacy. However, compared with single-agent therapies, combination immunotherapies are associated with increased overall toxicity because the very same mechanisms also work in concert to enhance systemic inflammation and promote off-tumor toxicity. Therefore, rational design of combination regimens that achieve improved antitumor control without exacerbated toxicity is a main objective in combination immunotherapy. Here, we show that the combination of engineered, tumor matrix-binding interleukin-7 (IL-7) and IL-12 achieves remarkable anticancer effects by activating complementary pathways without inducing any additive immunotoxicity. Mechanistically, engineered IL-12 provided effector properties to T cells, while IL-7 prevented their exhaustion and boosted memory formation as assessed by tumor rechallenge experiments. The dual combination also rendered checkpoint inhibitor (CPI)-resistant genetically engineered melanoma model responsive to CPI. Thus, our approach provides a framework of evaluation of rationally designed combinations in immuno-oncology and yields a promising therapy.


Subject(s)
Interleukin-12 , Melanoma , Humans , Interleukin-12/genetics , Interleukin-7/pharmacology , T-Cell Exhaustion , Immunotherapy , Melanoma/pathology
5.
NPJ Regen Med ; 8(1): 49, 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37696884

ABSTRACT

Non-healing wounds have a negative impact on quality of life and account for many cases of amputation and even early death among patients. Diabetic patients are the predominate population affected by these non-healing wounds. Despite the significant clinical demand, treatment with biologics has not broadly impacted clinical care. Interleukin-4 (IL-4) is a potent modulator of the immune system, capable of skewing macrophages towards a pro-regeneration phenotype (M2) and promoting angiogenesis, but can be toxic after frequent administration and is limited by its short half-life and low bioavailability. Here, we demonstrate the design and characterization of an engineered recombinant interleukin-4 construct. We utilize this collagen-binding, serum albumin-fused IL-4 variant (CBD-SA-IL-4) delivered in a hyaluronic acid (HA)-based gel for localized application of IL-4 to dermal wounds in a type 2 diabetic mouse model known for poor healing as proof-of-concept for improved tissue repair. Our studies indicate that CBD-SA-IL-4 is retained within the wound and can modulate the wound microenvironment through induction of M2 macrophages and angiogenesis. CBD-SA-IL-4 treatment significantly accelerated wound healing compared to native IL-4 and HA vehicle treatment without inducing systemic side effects. This CBD-SA-IL-4 construct can address the underlying immune dysfunction present in the non-healing wound, leading to more effective tissue healing in the clinic.

6.
ACS Cent Sci ; 8(10): 1435-1446, 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36313164

ABSTRACT

Immune stimulating agents like Toll-like receptor 7 (TLR7) agonists induce potent antitumor immunity but are limited in their therapeutic window due to off-target immune activation. Here, we developed a polymeric delivery platform that binds excess unpaired cysteines on tumor cell surfaces and debris to adjuvant tumor neoantigens as an in situ vaccine. The metabolic and enzymatic dysregulation in the tumor microenvironment produces these exofacial free thiols, which can undergo efficient disulfide exchange with thiol-reactive pyridyl disulfide moieties upon intratumoral injection. These functional monomers are incorporated into a copolymer with pendant mannose groups and TLR7 agonists to target both antigen and adjuvant to antigen presenting cells. When tethered in the tumor, the polymeric glyco-adjuvant induces a robust antitumor response and prolongs survival of tumor-bearing mice, including in checkpoint-resistant B16F10 melanoma. The construct additionally reduces systemic toxicity associated with clinically relevant small molecule TLR7 agonists.

7.
Nat Biomed Eng ; 6(7): 819-829, 2022 07.
Article in English | MEDLINE | ID: mdl-35534574

ABSTRACT

Immune-checkpoint inhibitors have shown modest efficacy against immunologically 'cold' tumours. Interleukin-12 (IL-12)-a cytokine that promotes the recruitment of immune cells into tumours as well as immune cell activation, also in cold tumours-can cause severe immune-related adverse events in patients. Here, by exploiting the preferential overexpression of proteases in tumours, we show that fusing a domain of the IL-12 receptor to IL-12 via a linker cleavable by tumour-associated proteases largely restricts the pro-inflammatory effects of IL-12 to tumour sites. In mouse models of subcutaneous adenocarcinoma and orthotopic melanoma, masked IL-12 delivered intravenously did not cause systemic IL-12 signalling and eliminated systemic immune-related adverse events, led to potent therapeutic effects via the remodelling of the immune-suppressive microenvironment, and rendered cold tumours responsive to immune-checkpoint inhibition. We also show that masked IL-12 is activated in tumour lysates from patients. Protease-sensitive masking of potent yet toxic cytokines may facilitate their clinical translation.


Subject(s)
Interleukin-12 , Melanoma , Animals , Cytokines , Immunotherapy , Interleukin-12/pharmacology , Mice , Peptide Hydrolases , Tumor Microenvironment
8.
Biomaterials ; 278: 121159, 2021 11.
Article in English | MEDLINE | ID: mdl-34634664

ABSTRACT

The SARS-CoV-2 virus has caused an unprecedented global crisis, and curtailing its spread requires an effective vaccine which elicits a diverse and robust immune response. We have previously shown that vaccines made of a polymeric glyco-adjuvant conjugated to an antigen were effective in triggering such a response in other disease models and hypothesized that the technology could be adapted to create an effective vaccine against SARS-CoV-2. The core of the vaccine platform is the copolymer p(Man-TLR7), composed of monomers with pendant mannose or a toll-like receptor 7 (TLR7) agonist. Thus, p(Man-TLR7) is designed to target relevant antigen-presenting cells (APCs) via mannose-binding receptors and then activate TLR7 upon endocytosis. The p(Man-TLR7) construct is amenable to conjugation to protein antigens such as the Spike protein of SARS-CoV-2, yielding Spike-p(Man-TLR7). Here, we demonstrate Spike-p(Man-TLR7) vaccination elicits robust antigen-specific cellular and humoral responses in mice. In adult and elderly wild-type mice, vaccination with Spike-p(Man-TLR7) generates high and long-lasting titers of anti-Spike IgGs, with neutralizing titers exceeding levels in convalescent human serum. Interestingly, adsorbing Spike-p(Man-TLR7) to the depot-forming adjuvant alum amplified the broadly neutralizing humoral responses to levels matching those in mice vaccinated with formulations based off of clinically-approved adjuvants. Additionally, we observed an increase in germinal center B cells, antigen-specific antibody secreting cells, activated T follicular helper cells, and polyfunctional Th1-cytokine producing CD4+ and CD8+ T cells. We conclude that Spike-p(Man-TLR7) is an attractive, next-generation subunit vaccine candidate, capable of inducing durable and robust antibody and T cell responses.


Subject(s)
COVID-19 , Immunity, Humoral , Adjuvants, Immunologic , Aged , Animals , Antibodies, Neutralizing , Antibodies, Viral , CD8-Positive T-Lymphocytes , COVID-19 Vaccines , Humans , Immunity, Cellular , Mice , SARS-CoV-2
9.
ACS Cent Sci ; 7(8): 1368-1380, 2021 Aug 25.
Article in English | MEDLINE | ID: mdl-34466656

ABSTRACT

The COVID-19 pandemic underscores the need for rapid, safe, and effective vaccines. In contrast to some traditional vaccines, nanoparticle-based subunit vaccines are particularly efficient in trafficking antigens to lymph nodes, where they induce potent immune cell activation. Here, we developed a strategy to decorate the surface of oxidation-sensitive polymersomes with multiple copies of the SARS-CoV-2 spike protein receptor-binding domain (RBD) to mimic the physical form of a virus particle. We evaluated the vaccination efficacy of these surface-decorated polymersomes (RBDsurf) in mice compared to RBD-encapsulated polymersomes (RBDencap) and unformulated RBD (RBDfree), using monophosphoryl-lipid-A-encapsulated polymersomes (MPLA PS) as an adjuvant. While all three groups produced high titers of RBD-specific IgG, only RBDsurf elicited a neutralizing antibody response to SARS-CoV-2 comparable to that of human convalescent plasma. Moreover, RBDsurf was the only group to significantly increase the proportion of RBD-specific germinal center B cells in the vaccination-site draining lymph nodes. Both RBDsurf and RBDencap drove similarly robust CD4+ and CD8+ T cell responses that produced multiple Th1-type cytokines. We conclude that a multivalent surface display of spike RBD on polymersomes promotes a potent neutralizing antibody response to SARS-CoV-2, while both antigen formulations promote robust T cell immunity.

10.
Am J Physiol Cell Physiol ; 321(2): C369-C383, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34232748

ABSTRACT

Since the discovery of cytokines, much effort has been put forth to achieve therapeutic translation for treatment of various diseases, including cancer and autoimmune diseases. Despite these efforts, very few cytokines have cleared regulatory approval, and those that were approved are not commonly used due to their challenging toxicity profile and/or limited therapeutic efficacy. The main limitation in translation has been that wild-type cytokines have unfavorable pharmacokinetic and pharmacodynamic profiles, either eliciting unwanted systemic side effects or insufficient residence in secondary lymphoid organs. In this review, we address protein-engineering approaches that have been applied to both proinflammatory and anti-inflammatory cytokines to enhance their therapeutic indices, and we highlight diseases in which administration of engineered cytokines is especially relevant.


Subject(s)
Cytokines/therapeutic use , Immunotherapy , Neoplasms/therapy , Protein Engineering , Animals , Cytokines/genetics , Drug Delivery Systems/methods , Humans , Immunotherapy/methods , Inflammation/drug therapy , Neoplasms/genetics , Neoplasms/immunology
11.
bioRxiv ; 2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33851166

ABSTRACT

A diverse portfolio of SARS-CoV-2 vaccine candidates is needed to combat the evolving COVID-19 pandemic. Here, we developed a subunit nanovaccine by conjugating SARS-CoV-2 Spike protein receptor binding domain (RBD) to the surface of oxidation-sensitive polymersomes. We evaluated the humoral and cellular responses of mice immunized with these surface-decorated polymersomes (RBDsurf) compared to RBD-encapsulated polymersomes (RBDencap) and unformulated RBD (RBDfree), using monophosphoryl lipid A-encapsulated polymersomes (MPLA PS) as an adjuvant. While all three groups produced high titers of RBD-specific IgG, only RBDsurf elicited a neutralizing antibody response to SARS-CoV-2 comparable to that of human convalescent plasma. Moreover, RBDsurf was the only group to significantly increase the proportion of RBD-specific germinal center B cells in the vaccination-site draining lymph nodes. Both RBDsurf and RBDencap drove similarly robust CD4+ and CD8+ T cell responses that produced multiple Th1-type cytokines. We conclude that multivalent surface display of Spike RBD on polymersomes promotes a potent neutralizing antibody response to SARS-CoV-2, while both antigen formulations promote robust T cell immunity.

12.
Nat Biomed Eng ; 5(5): 387-398, 2021 05.
Article in English | MEDLINE | ID: mdl-33046864

ABSTRACT

Interleukin-4 (IL-4) suppresses the development of multiple sclerosis in a murine model of experimental autoimmune encephalomyelitis (EAE). Here, we show that, in mice with EAE, the accumulation and persistence in the lymph nodes and spleen of a systemically administered serum albumin (SA)-IL-4 fusion protein leads to higher efficacy in preventing disease development than the administration of wild-type IL-4 or of the clinically approved drug fingolimod. We also show that the SA-IL-4 fusion protein prevents immune-cell infiltration in the spinal cord, decreases integrin expression in antigen-specific CD4+ T cells, increases the number of granulocyte-like myeloid-derived suppressor cells (and their expression of programmed-death-ligand-1) in spinal cord-draining lymph nodes and decreases the number of T helper 17 cells, a pathogenic cell population in EAE. In mice with chronic EAE, SA-IL-4 inhibits immune-cell infiltration into the spinal cord and completely abrogates immune responses to myelin antigen in the spleen. The SA-IL-4 fusion protein may be prophylactically and therapeutically advantageous in the treatment of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunosuppressive Agents/administration & dosage , Interleukin-4/metabolism , Recombinant Fusion Proteins/administration & dosage , Serum Albumin/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Half-Life , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/pharmacology , Injections, Intravenous , Lymph Nodes/chemistry , Lymph Nodes/immunology , Mice , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacology , Spleen/chemistry , Spleen/immunology , Th17 Cells/drug effects
13.
Arthritis Rheumatol ; 73(5): 769-778, 2021 05.
Article in English | MEDLINE | ID: mdl-33169522

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) is a major autoimmune disease that causes synovitis and joint damage. Although clinical trials have been performed using interleukin-10 (IL-10), an antiinflammatory cytokine, as a potential treatment of RA, the therapeutic effects of IL-10 have been limited, potentially due to insufficient residence in lymphoid organs, where antigen recognition primarily occurs. This study was undertaken to engineer an IL-10-serum albumin (SA) fusion protein and evaluate its effects in 2 murine models of RA. METHODS: SA-fused IL-10 (SA-IL-10) was recombinantly expressed. Mice with collagen antibody-induced arthritis (n = 4-7 per group) or collagen-induced arthritis (n = 9-15 per group) were injected intravenously with wild-type IL-10 or SA-IL-10, and the retention of SA-IL-10 in the lymph nodes (LNs), immune cell composition in the paws, and therapeutic effect of SA-IL-10 on mice with arthritis were assessed. RESULTS: SA fusion to IL-10 led to enhanced accumulation in the mouse LNs compared with unmodified IL-10. Intravenous SA-IL-10 treatment restored immune cell composition in the paws to a normal status, elevated the frequency of suppressive alternatively activated macrophages, reduced IL-17A levels in the paw-draining LN, and protected joint morphology. Intravenous SA-IL-10 treatment showed similar efficacy as treatment with an anti-tumor necrosis factor antibody. SA-IL-10 was equally effective when administered intravenously, locally, or subcutaneously, which is a benefit for clinical translation of this molecule. CONCLUSION: SA fusion to IL-10 is a simple but effective engineering strategy for RA therapy and has potential for clinical translation.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Foot Joints/drug effects , Interleukin-10/pharmacology , Lymph Nodes/immunology , Macrophages/drug effects , Recombinant Fusion Proteins/pharmacology , Serum Albumin/pharmacology , Animals , Antigen-Presenting Cells/metabolism , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Disease Models, Animal , Foot , Foot Joints/immunology , Foot Joints/metabolism , Foot Joints/pathology , Hindlimb , Histocompatibility Antigens Class I/metabolism , Injections, Intravenous , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-6/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/immunology , Mice , Protein Engineering , Protein Transport , Receptors, Fc/metabolism , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor Inhibitors/pharmacology
15.
Nat Biomed Eng ; 4(5): 531-543, 2020 05.
Article in English | MEDLINE | ID: mdl-32284554

ABSTRACT

Checkpoint-inhibitor (CPI) immunotherapy has achieved remarkable clinical success, yet its efficacy in 'immunologically cold' tumours has been modest. Interleukin-12 (IL-12) is a powerful cytokine that activates the innate and adaptive arms of the immune system; however, the administration of IL-12 has been associated with immune-related adverse events. Here we show that, after intravenous administration of a collagen-binding domain fused to IL-12 (CBD-IL-12) in mice bearing aggressive mouse tumours, CBD-IL-12 accumulates in the tumour stroma due to exposed collagen in the disordered tumour vasculature. In comparison with the administration of unmodified IL-12, CBD-IL-12 induced sustained intratumoural levels of interferon-γ, substantially reduced its systemic levels as well as organ damage and provided superior anticancer efficacy, eliciting complete regression of CPI-unresponsive breast tumours. Furthermore, CBD-IL-12 potently synergized with CPI to eradicate large established melanomas, induced antigen-specific immunological memory and controlled tumour growth in a genetically engineered mouse model of melanoma. CBD-IL-12 may potentiate CPI immunotherapy for immunologically cold tumours.


Subject(s)
Collagen/metabolism , Inflammation/pathology , Interleukin-12/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Adaptive Immunity/drug effects , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Models, Animal , Epitopes/immunology , Female , Immunity, Innate/drug effects , Interleukin-12/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Protein Binding/drug effects , Protein Domains , Remission Induction , Tumor Burden/drug effects , Tumor Microenvironment/drug effects
16.
Sci Adv ; 5(12): eaay1357, 2019 12.
Article in English | MEDLINE | ID: mdl-31844672

ABSTRACT

Although a clinical breakthrough for cancer treatment, it remains that a minority of patients respond to checkpoint inhibitor (CPI) immunotherapy. The composition of tumor-infiltrating immune cells has been identified as a key factor influencing CPI therapy success. Thus, enhancing tumor immune cell infiltration is a critical challenge. A lack of the chemokine CCL4 within the tumor microenvironment leads to the absence of CD103+ dendritic cells (DCs), a crucial cell population influencing CPI responsiveness. Here, we use a tumor stroma-targeting approach to deliver CCL4; by generating a fusion protein of CCL4 and the collagen-binding domain (CBD) of von Willebrand factor, we show that CBD fusion enhances CCL4 tumor localization. Intravenous CBD-CCL4 administration recruits CD103+ DCs and CD8+ T cells and improves the antitumor effect of CPI immunotherapy in multiple tumor models, including poor responders to CPI. Thus, CBD-CCL4 holds clinical translational potential by enhancing efficacy of CPI immunotherapy.


Subject(s)
Antigens, CD/immunology , Chemokine CCL4/genetics , Immunotherapy , Integrin alpha Chains/immunology , Melanoma, Experimental/immunology , Melanoma/immunology , Animals , Antigens, CD/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/immunology , Chemokine CCL4/immunology , Chemokine CCL4/pharmacology , Collagen/genetics , Collagen/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Integrin alpha Chains/genetics , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/genetics , Melanoma/therapy , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Protein Binding/genetics , Tumor Microenvironment/drug effects , von Willebrand Factor/genetics
17.
Sci Adv ; 5(11): eaay1971, 2019 11.
Article in English | MEDLINE | ID: mdl-31723606

ABSTRACT

Enhancing the therapeutic efficacy of drugs for inflammatory diseases is of high demand. One possible approach is targeting drugs to the extracellular matrix of the inflamed area. Here, we target collagens in the matrix, which are inaccessible in most tissues yet are exposed to the bloodstream in the inflamed area because of vascular hyperpermeability. We conferred collagen affinity to anti-tumor necrosis factor-α (α-TNF) antibody by conjugating a collagen-binding peptide (CBP) derived from the sequence of decorin. CBP-α-TNF accumulated in the inflamed paw of the arthritis model, and arthritis development was significantly suppressed by treatment with CBP-α-TNF compared with the unmodified antibody. Similarly, CBP-anti-transforming growth factor-ß (α-TGF-ß) accumulated in the inflamed lung of pulmonary fibrosis model and significantly suppressed pulmonary fibrosis compared with the unmodified antibody. Together, collagen affinity enables the anticytokine antibodies to target arthritis and pulmonary fibrosis accompanied by inflammation, demonstrating a clinically translational approach to treat inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies/therapeutic use , Collagen/antagonists & inhibitors , Inflammation/drug therapy , Lung/drug effects , Pulmonary Fibrosis/drug therapy , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Antibodies/immunology , Antibodies/metabolism , Collagen/immunology , Collagen/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Lung/immunology , Lung/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Peptide Fragments/immunology , Peptide Fragments/metabolism , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/metabolism , Sialoglycoproteins/immunology , Sialoglycoproteins/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
18.
Sci Adv ; 5(8): eaaw6081, 2019 08.
Article in English | MEDLINE | ID: mdl-31453327

ABSTRACT

Serum albumin (SA) is used as a carrier to deliver cytotoxic agents to tumors via passive targeting. To further improve SA's tumor targeting capacity, we sought to develop an approach to retain SA-drug conjugates within tumors through a combination of passive and active targeting. SA was recombinantly fused with a collagen-binding domain (CBD) of von Willebrand factor to bind within the tumor stroma after extravasation due to tumor vascular permeability. Doxorubicin (Dox) was conjugated to the CBD-SA via a pH-sensitive linker. Dox-CBD-SA treatment significantly suppressed tumor growth compared to both Dox-SA and aldoxorubicin treatment in a mouse model of breast cancer. Dox-CBD-SA efficiently stimulated host antitumor immunity, resulting in the complete eradication of MC38 colon carcinoma when used in combination with anti-PD-1 checkpoint inhibitor. Dox-CBD-SA decreased adverse events compared to aldoxorubicin. Thus, engineered CBD-SA could be a versatile and clinically relevant drug conjugate carrier protein for treatment of solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Doxorubicin/pharmacology , Drug Carriers/chemistry , Serum Albumin/chemistry , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Doxorubicin/chemistry , Female , Hydrazones/pharmacology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Domains , von Willebrand Factor/chemistry
19.
Sci Transl Med ; 11(487)2019 04 10.
Article in English | MEDLINE | ID: mdl-30971453

ABSTRACT

Cancer immunotherapy with immune checkpoint inhibitors (CPIs) and interleukin-2 (IL-2) has demonstrated clinical efficacy but is frequently accompanied with severe adverse events caused by excessive and systemic immune system activation. Here, we addressed this need by targeting both the CPI antibodies anti-cytotoxic T lymphocyte antigen 4 antibody (αCTLA4) + anti-programmed death ligand 1 antibody (αPD-L1) and the cytokine IL-2 to tumors via conjugation (for the antibodies) or recombinant fusion (for the cytokine) to a collagen-binding domain (CBD) derived from the blood protein von Willebrand factor (VWF) A3 domain, harnessing the exposure of tumor stroma collagen to blood components due to the leakiness of the tumor vasculature. We show that intravenously administered CBD protein accumulated mainly in tumors. CBD conjugation or fusion decreases the systemic toxicity of both αCTLA4 + αPD-L1 combination therapy and IL-2, for example, eliminating hepatotoxicity with the CPI molecules and ameliorating pulmonary edema with IL-2. Both CBD-CPI and CBD-IL-2 suppressed tumor growth compared to their unmodified forms in multiple murine cancer models, and both CBD-CPI and CBD-IL-2 increased tumor-infiltrating CD8+ T cells. In an orthotopic breast cancer model, combination treatment with CPI and IL-2 eradicated tumors in 9 of 13 animals with the CBD-modified drugs, whereas it did so in only 1 of 13 animals with the unmodified drugs. Thus, the A3 domain of VWF can be used to improve safety and efficacy of systemically administered tumor drugs with high translational promise.


Subject(s)
Antibodies, Neoplasm/immunology , Collagen/metabolism , Cytokines/immunology , Immunotherapy , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytotoxicity, Immunologic , Disease Models, Animal , Humans , Immunity , Injections, Intravenous , Interleukin-2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Domains , Treatment Outcome
20.
Environ Sci Technol ; 52(1): 162-172, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29182855

ABSTRACT

Insight into the mechanisms underlying bacterial adhesion is critical to the formulation of membrane biofouling control strategies. Using AFM-based single-cell force spectroscopy, we investigated the interaction between Pseudomonas fluorescens, a biofilm-forming bacterium, and polysulfone (PSF) ultrafiltration (UF) membranes to unravel the mechanisms underlying early stage membrane biofouling. We show that hydrophilic polydopamine (PDA) coatings decrease bacterial adhesion forces at short bacterium-membrane contact times. Further, we find that adhesion forces are weakened by the presence of natural organic matter (NOM) conditioning films, owing to the hydrophilicity of NOM. Investigation of the effect of adhesion contact time revealed that PDA coatings are less effective at preventing bioadhesion when the contact time is prolonged to 2-5 s, or when the membranes are exposed to bacterial suspensions under stirring. These results therefore challenge the notion that simple hydrophilic surface coatings are effective as a biofouling control strategy. Finally, we present evidence that adhesion to the UF membrane surface is mediated by cell-surface macromolecules (likely to be outer membrane proteins and pili) which, upon contacting the membrane, undergo surface-induced unfolding.


Subject(s)
Biofouling , Ultrafiltration , Bacterial Adhesion , Hydrophobic and Hydrophilic Interactions , Membranes, Artificial
SELECTION OF CITATIONS
SEARCH DETAIL
...