Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 10(5): 626-640, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35319751

ABSTRACT

The CD40 receptor is an attractive target for cancer immunotherapy. Although a modest pharmacodynamic effect is seen in patients following administration of CD40-targeting monoclonal antibodies (mAb), the doses that could be safely administered do not result in a meaningful clinical response, most likely due to the limited therapeutic window associated with systemic CD40 activation. To overcome this issue, we developed a multispecific DARPin construct, α-FAPxCD40, which has conditional activity at the site of disease. α-FAPxCD40 activation of CD40 depends on binding to fibroblast activation protein (FAP), a cell-surface protease overexpressed in the stroma of solid tumors. In vitro studies demonstrated that α-FAPxCD40 potently activates human antigen-presenting cells in the presence, but not in the absence, of FAP-positive cells. After intravenous injection, a murine surrogate construct (α-mFAPxCD40) accumulated in FAP-positive tumors, elicited rejection of 88% of these tumors, and induced memory antitumor immunity. Importantly, in contrast to the mouse anti-CD40 tested in parallel, the in vivo antitumor activity of α-mFAPxCD40 was associated neither with elevated blood cytokines nor with hepatotoxicity, both of which contribute to the clinical dose-limiting toxicities of several CD40 mAb. This study demonstrates that α-(m)FAPxCD40 engages CD40 in an FAP-restricted manner, leading to tumor eradication without signs of peripheral toxicity. This distinct preclinical profile suggests that a favorable therapeutic index may be achieved in humans. It further supports the development of α-FAPxCD40, currently tested in a first-in-human clinical study in patients with solid tumors (NCT05098405).


Subject(s)
Antineoplastic Agents, Immunological , Neoplasms , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , CD40 Antigens , Cell Line, Tumor , Designed Ankyrin Repeat Proteins , Humans , Immunotherapy , Lymphocyte Activation , Mice , Neoplasms/drug therapy
2.
Proc Natl Acad Sci U S A ; 117(1): 541-551, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31889004

ABSTRACT

Cancer immunotherapies are increasingly combined with targeted therapies to improve therapeutic outcomes. We show that combination of agonistic anti-CD40 with antiangiogenic antibodies targeting 2 proangiogenic factors, vascular endothelial growth factor A (VEGFA) and angiopoietin 2 (Ang2/ANGPT2), induces pleiotropic immune mechanisms that facilitate tumor rejection in several tumor models. On the one hand, VEGFA/Ang2 blockade induced regression of the tumor microvasculature while decreasing the proportion of nonperfused vessels and reducing leakiness of the remaining vessels. On the other hand, both anti-VEGFA/Ang2 and anti-CD40 independently promoted proinflammatory macrophage skewing and increased dendritic cell activation in the tumor microenvironment, which were further amplified upon combination of the 2 treatments. Finally, combined therapy provoked brisk infiltration and intratumoral redistribution of cytotoxic CD8+ T cells in the tumors, which was mainly driven by Ang2 blockade. Overall, these nonredundant synergistic mechanisms endowed T cells with improved effector functions that were conducive to more efficient tumor control, underscoring the therapeutic potential of antiangiogenic immunotherapy in cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , CD40 Antigens/agonists , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment/drug effects , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD40 Antigens/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Synergism , Female , Humans , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasms/blood supply , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
3.
Sci Transl Med ; 9(385)2017 04 12.
Article in English | MEDLINE | ID: mdl-28404865

ABSTRACT

Pathological angiogenesis is a hallmark of cancer and a therapeutic target. Vascular endothelial growth factor A (VEGFA) and angiopoietin-2 (ANGPT2; also known as ANG2) are proangiogenic cytokines that sustain tumor angiogenesis and limit antitumor immunity. We show that combined ANGPT2 and VEGFA blockade by a bispecific antibody (A2V) provided superior therapeutic benefits, as compared to the single agents, in both genetically engineered and transplant tumor models, including metastatic breast cancer (MMTV-PyMT), pancreatic neuroendocrine tumor (RIP1-Tag2), and melanoma. Mechanistically, A2V promoted vascular regression, tumor necrosis, and antigen presentation by intratumoral phagocytes. A2V also normalized the remaining blood vessels and facilitated the extravasation and perivascular accumulation of activated, interferon-γ (IFNγ)-expressing CD8+ cytotoxic T lymphocytes (CTLs). Whereas the antitumoral activity of A2V was, at least partly, CTL-dependent, perivascular T cells concurrently up-regulated the expression of the immune checkpoint ligand programmed cell death ligand 1 (PD-L1) in tumor endothelial cells. IFNγ neutralization blunted this adaptive response, and PD-1 blockade improved tumor control by A2V in different cancer models. These findings position immune cells as key effectors of antiangiogenic therapy and support the rationale for cotargeting angiogenesis and immune checkpoints in cancer therapy.


Subject(s)
Angiopoietin-2/metabolism , Programmed Cell Death 1 Receptor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiopoietin-2/genetics , Animals , B7-H1 Antigen/metabolism , Blood Vessels/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Mice , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , Vascular Endothelial Growth Factor A/genetics
4.
Cell Rep ; 8(3): 696-706, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25088418

ABSTRACT

Angiopoietin-2 (ANG2/ANGPT2) is a context-dependent TIE2 receptor agonist/antagonist and proangiogenic factor. Although ANG2 neutralization improves tumor angiogenesis and growth inhibition by vascular endothelial growth factor (VEGF)-A signaling blockade, the mechanistic underpinnings of such therapeutic benefits remain poorly explored. We employed late-stage RIP1-Tag2 pancreatic neuroendocrine tumors (PNETs) and MMTV-PyMT mammary adenocarcinomas, which develop resistance to VEGF receptor 2 (VEGFR2) blockade. We found that VEGFR2 inhibition upregulated ANG2 and vascular TIE2 and enhanced infiltration by TIE2-expressing macrophages in the PNETs. Dual ANG2/VEGFR2 blockade suppressed revascularization and progression in most of the PNETs, whereas it had only minor additive effects in the mammary tumors, which did not upregulate ANG2 upon VEGFR2 inhibition. ANG2/VEGFR2 blockade did not elicit increased PNET invasion and metastasis, although it exacerbated tumor hypoxia and hematopoietic cell infiltration. These findings suggest that evasive tumor resistance to anti-VEGFA therapy may involve the adaptive enforcement of ANG2-TIE2 signaling, which can be reversed by ANG2 neutralization.


Subject(s)
Adenocarcinoma/metabolism , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Pancreatic Neoplasms/metabolism , Ribonuclease, Pancreatic/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adenocarcinoma/drug therapy , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/drug therapy , Ribonuclease, Pancreatic/antagonists & inhibitors , Ribonuclease, Pancreatic/immunology , Signal Transduction , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/immunology
5.
Cell Rep ; 4(4): 621-3, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23993444

ABSTRACT

Angiopoietin-2 (ANG2/ANGPT2) has recently emerged as a promising target for the inhibition of tumor metastasis. In this issue of Cell Reports, Minami et al. show that the calcineurin-NFAT pathway promotes pulmonary tumor metastasis by inducing ANG2 in the lung endothelium.


Subject(s)
Calcineurin/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , NFATC Transcription Factors/metabolism , Ribonuclease, Pancreatic/metabolism , Animals , Humans
7.
Cancer Immunol Immunother ; 61(4): 453-68, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22331081

ABSTRACT

The recent FDA approval of the first therapeutic vaccine against prostate cancer has revitalized the public interest in the fields of cancer immunology and immunotherapy. Yet, clinical results are modest. A reason for this limited success may reside in the capacity of the tumor to convert inflammation in a tumor-promoting condition and eventually escape immune surveillance. Here we present the main known interactions between the prostate tumor and the immune system, showing how the malignancy can dodge the immune system by also exerting several immunosuppressive mechanisms. We also discuss experimental and clinical strategies proposed to counteract cancer immune evasion and emphasize the importance of implementing appropriate murine models like the transgenic adenocarcinoma of the mouse prostate model for investigating the biology of prostate cancer and novel immunotherapy approaches against it.


Subject(s)
Adenocarcinoma/immunology , Adenocarcinoma/therapy , Cancer Vaccines/therapeutic use , Immunotherapy/trends , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Adenocarcinoma/pathology , Animals , Disease Models, Animal , Humans , Immunologic Surveillance , Immunosuppression Therapy , Male , Mice , Prostatic Neoplasms/pathology , Tumor Escape
8.
Clin Cancer Res ; 17(5): 1012-23, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21248302

ABSTRACT

PURPOSE: Chronic inflammation, recruitment of myeloid-derived cells, and perturbation of the arginine metabolism have been all proposed as mechanisms favoring prostate carcinogenesis and tumor immunoescape. Objective of this study was to evaluate whether accumulation of CD11b(+)Gr1(+) cells, also defined myeloid-derived suppressor cells, occur in mice affected by transplantable or spontaneous prostate cancer (PC). We also investigated whether N(G) nitro-L-arginine methyl ester (L-NAME) and sildenafil, both modulators of the arginine metabolism, restrain tumor growth and restore tumor-specific immunity. EXPERIMENTAL DESIGN: Wild-type C57BL/6 mice bearing TRAMP-C1 PC and transgenic adenocarcinoma of the mouse prostate (TRAMP) mice were treated with vehicle, L-NAME or sildenafil, and evaluated for CD11b(+) cells accumulation in the blood, several organs, and the tumor mass and for disease progression. RESULTS: CD11b(+)Gr1(high), CD11b(+)Gr1(int), and CD11b(+)Gr1(-) cells differently accumulated in different organs and especially in the tumor of the two mouse models. L-NAME and sildenafil impaired the immunosuppressive function of CD11b(+) cells in both models and restrained TRAMP-C1 growth, but they neither break tumor-specific immune tolerance nor limit tumor progression in TRAMP mice. CONCLUSIONS: Collectively, our results emphasize substantial differences in tumor-induced alteration of myelopoiesis and sensitivity to modulators of the arginine metabolism between a transplantable and a spontaneous model of PC. They also suggest that perturbation of the arginine metabolism is dispensable for PC progression and the associated T-cell tolerance.


Subject(s)
Arginine/metabolism , NG-Nitroarginine Methyl Ester/pharmacology , Piperazines/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Sulfones/pharmacology , T-Lymphocytes/immunology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Animals , CD11b Antigen , Cell Proliferation/drug effects , Disease Progression , Immune Tolerance/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelopoiesis , Prostatic Neoplasms/metabolism , Purines/pharmacology , Sildenafil Citrate , Tumor Cells, Cultured
9.
N Biotechnol ; 25(6): 417-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19552886

ABSTRACT

Abnormal activity of the epidermal growth factor receptor (EGFR) is associated with various cancer-related processes and motivates the search for strategies that can selectively block EGFR signalling. In this study, functional knockdown of EGFR was achieved through expression of an affibody construct, (ZEGFR:1907)(2-)KDEL, with high affinity for EGFR and extended with the amino acids KDEL to make it resident in the secretory compartments. Expression of (ZEGFR:1907)(2-)KDEL resulted in 80% reduction ofthe cell surface level of EGFR, and fluorescent staining for EGFR and the (ZEGFR:1907)(2-)KDEL construct showed overlapping intracellular localisation. Immunocapture of EGFR from cell lysates showed that an intracellular complex between EGFR and the affibody construct had been formed, further indicating aspecific interaction between the affibody construct and EGFR. Surface depletion of EGFR led to a dramatic decrease in the amount of kinase domain phosphorylated EGFR, coincident with a significant decrease in the proliferation rate.


Subject(s)
Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Phosphotransferases/metabolism , Recombinant Fusion Proteins/metabolism , Secretory Vesicles/metabolism , Cell Line, Tumor , Humans , Phosphorylation , Protein Structure, Tertiary
10.
BMC Immunol ; 10: 1, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19134173

ABSTRACT

BACKGROUND: Tumor-associated accrual of myeloid derived suppressor cells (MDSC) in the blood, lymphoid organs and tumor tissues may lead to perturbation of the arginine metabolism and impairment of the endogenous antitumor immunity. The objective of this study was to evaluate whether accumulation of MDSC occurred in Th2 prone BALB/c and Th1 biased C57BL/6 mice bearing the C26GM colon carcinoma and RMA T lymphoma, respectively, and to investigate whether N(G) nitro-L-arginine methyl ester (L-NAME) and sildenafil, both modulators of the arginine metabolism, restored antitumor immunity. RESULTS: We report here that MDSC accumulate in the spleen and blood of mice irrespective of the mouse and tumor model used. Treatment of tumor-bearing mice with either the phosphodiesterase-5 inhibitor sildenafil or the nitric-oxide synthase (NOS) inhibitor L-NAME significantly restrained tumor growth and expanded the tumor-specific immune response. CONCLUSION: Our data emphasize the role of MDSC in modulating the endogenous tumor-specific immune response and underline the anti-neoplastic therapeutic potential of arginine metabolism modulators.


Subject(s)
Arginine/metabolism , Immunologic Factors/metabolism , Immunologic Surveillance , Neoplasms/immunology , Animals , Arginine/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Immunologic Surveillance/drug effects , Interferon-gamma/metabolism , Lymph Nodes/drug effects , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/pathology , Myelopoiesis/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Nitric Oxide Synthase Type II/antagonists & inhibitors , Spleen/drug effects , Spleen/pathology , Weight Loss/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...