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










Publication year range
1.
Nat Rev Clin Oncol ; 21(6): 468-482, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38600370

ABSTRACT

Antiangiogenic agents, generally antibodies or tyrosine-kinase inhibitors that target the VEGF-VEGFR pathway, are currently among the few combination partners clinically proven to improve the efficacy of immune-checkpoint inhibitors (ICIs). This benefit has been demonstrated in pivotal phase III trials across different cancer types, some with practice-changing results; however, numerous phase III trials have also had negative results. The rationale for using antiangiogenic drugs as partners for ICIs relies primarily on blocking the multiple immunosuppressive effects of VEGF and inducing several different vascular-modulating effects that can stimulate immunity, such as vascular normalization leading to increased intratumoural blood perfusion and flow, and inhibition of pro-apoptotic effects of endothelial cells on T cells, among others. Conversely, VEGF blockade can also cause changes that suppress antitumour immunity, such as increased tumour hypoxia, and reduced intratumoural ingress of co-administered ICIs. As a result, the net clinical benefits from antiangiogenic-ICI combinations will be determined by the balance between the opposing effects of VEGF signalling and its inhibition on the antitumour immune response. In this Perspective, we summarize the results from the currently completed phase III trials evaluating antiangiogenic agent-ICI combinations. We also discuss strategies to improve the efficacy of these combinations, focusing on aspects that include the deleterious functions of VEGF-VEGFR inhibition on antitumour immunity, vessel co-option as a driver of non-angiogenic tumour growth, clinical trial design, or the rationale for drug selection, dosing and scheduling.


Subject(s)
Angiogenesis Inhibitors , Immune Checkpoint Inhibitors , Neoplasms , Humans , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials, Phase III as Topic , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors
2.
Sci Immunol ; 7(67): eabm6388, 2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35030032

ABSTRACT

Tumors can escape immunity through multiple mechanisms, one of which is by enforcing a state of unresponsiveness of the tumor vasculature to inflammatory cytokines. This results in a lack of adhesiveness of angiogenic endothelial cells for immune cells and thus compromised immunity. This type of escape from immunity, called tumor endothelial cell anergy, is the result of exposure to angiogenic growth factors. Angiogenesis is a hallmark not only of cancer but also of embryonic development. It is assumed that angiogenesis-induced suppression of adhesion molecules is a regulatory function to provide an embryo with immune privileged conditions and allow uninterrupted growth and development. It is becoming clear that similar conditions are used by tumors to evade the immune system and ensure progressive growth. Gaining enhanced insight into these immune-privileged conditions is important as endothelial cell anergy can be overcome by angiogenesis inhibitors, an application that is rapidly emerging as a successful strategy to improve immunotherapy. The literature on endothelial adhesion molecule expression and leukocyte-vessel wall interactions during embryonic and fetal development is sparse, but available data allow the hypothesis that tumors, through angiogenesis, enforce an embryonic-like gene expression program in endothelial cells to suppress leukocyte infiltration and compromise antitumor immunity.


Subject(s)
Endothelial Cells/immunology , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Animals , Humans , Immunotherapy , Neoplasms/therapy , Neovascularization, Pathologic/therapy
3.
EMBO Mol Med ; 13(7): e08253, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34125494

ABSTRACT

Anti-angiogenic drugs targeting the VEGF pathway are most effective in advanced metastatic disease settings of certain types of cancers, whereas they have been unsuccessful as adjuvant therapies of micrometastatic disease in numerous phase III trials involving early-stage (resectable) cancers. Newer investigational anti-angiogenic drugs have been designed to inhibit the Angiopoietin (Ang)-Tie pathway. Acting through Tie2 receptors, the Ang1 ligand is a gatekeeper of endothelial quiescence. Ang2 is a dynamically expressed pro-angiogenic destabilizer of endothelium, and its upregulation is associated with poor prognosis in cancer. Besides using Ang2 blockers as inhibitors of tumor angiogenesis, little attention has been paid to their use as stabilizers of blood vessels to suppress tumor cell extravasation and metastasis. In clinical trials, Ang2 blockers have shown limited efficacy in advanced metastatic disease settings. This review summarizes preclinical evidence suggesting the potential utility of Ang2 inhibitors or Tie2 activators as neoadjuvant or adjuvant therapies in the prevention or treatment of early-stage micrometastatic disease. We further discuss the rationale and potential of combining these strategies with immunotherapy, including immune checkpoint targeting antibodies.


Subject(s)
Angiopoietin-2 , Neoplasms , Angiogenesis Inhibitors/therapeutic use , Angiopoietin-1 , Humans , Immunotherapy , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/prevention & control , Receptor, TIE-2
4.
NPJ Breast Cancer ; 6: 29, 2020.
Article in English | MEDLINE | ID: mdl-32704531

ABSTRACT

The impressive successes of immune checkpoint blockade antibodies to treat various types of cancer are limited to minor subsets of patients. Combination therapy strategies, including with chemotherapy, are being explored to possibly improve the efficacy of immunotherapies. Here we report results regarding the use of an immunostimulatory regimen of metronomic cyclophosphamide (CTX). We show that in orthotopic models of syngeneic murine triple-negative breast cancer (EMT6), CTX administered at 140 mg/kg every 6 days (CTX140 1q6d) is superior at inhibiting primary tumor growth when compared to maximum tolerated dose or daily oral (continuous) low-dose CTX. In SCID or SCID beige mice, anti-tumor effects of CTX140 1q6d are reduced, reinforcing the therapeutic contribution of the adaptive and innate immune systems. In a second breast cancer model (SP1-AC2M2), CTX140 1q6d again showed clear superiority in anti-tumor effects, causing complete tumor regressions; however, these mice were not protected from subsequent tumor re-challenge, suggesting absence of immune memory. We also show that in an aggressive and metastatic cisplatin-resistant variant (EMT6-CDDP), CTX140 1q6d is superior and invokes an influx of intra-tumoral CD4+ and CD8+ T cells. CTX increases expression of tumor cell PD-L1; however, when combined with concomitant PD-L1 antibody therapy none of the CTX regimens showed increased benefit. This work sheds light on the potential use of metronomic CTX for the treatment of breast cancer, in particular using the quasi-weekly regimen, but also underscores the complexity of the anti-tumor mechanisms and potential to improve immune checkpoint therapy efficacy.

5.
EMBO Mol Med ; 12(6): e12355, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32406209

ABSTRACT

Targeting the metastatic process is a critical pursuit in the treatment of malignant disease. There are currently no specific anti-metastatic drugs approved for clinical use, despite metastasis being the leading cause of death for cancer patients. Targeting the Tie1 receptor was shown as a possible strategy for selective anti-metastasis therapies based on previous gene deletion studies. This current study is the first description of a human antibody against Tie1 with the potential for clinical use in targeting extravasation of tumor cells into organs such as the lung, without having a detrimental effect on immune cell infiltration.


Subject(s)
Neoplasms , Antibodies , Humans , Neoplasm Metastasis , Neoplasms/drug therapy
6.
FEBS J ; 286(17): 3299-3332, 2019 09.
Article in English | MEDLINE | ID: mdl-31287944

ABSTRACT

The C-type lectin domain (CTLD) group 14 family of transmembrane glycoproteins consist of thrombomodulin, CD93, CLEC14A and CD248 (endosialin or tumour endothelial marker-1). These cell surface proteins exhibit similar ectodomain architecture and yet mediate a diverse range of cellular functions, including but not restricted to angiogenesis, inflammation and cell adhesion. Thrombomodulin, CD93 and CLEC14A can be expressed by endothelial cells, whereas CD248 is expressed by vasculature associated pericytes, activated fibroblasts and tumour cells among other cell types. In this article, we review the current literature of these family members including their expression profiles, interacting partners, as well as established and speculated functions. We focus primarily on their roles in the vasculature and inflammation as well as their contributions to tumour immunology. The CTLD group 14 family shares several characteristic features including their ability to be proteolytically cleaved and engagement of some shared extracellular matrix ligands. Each family member has strong links to tumour development and in particular CD93, CLEC14A and CD248 have been proposed as attractive candidate targets for cancer therapy.


Subject(s)
Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Receptors, Complement/metabolism , Thrombomodulin/metabolism , Animals , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Humans , Lectins, C-Type/genetics , Membrane Glycoproteins/genetics , Neoplasms/metabolism , Neovascularization, Physiologic , Receptors, Complement/genetics , Thrombomodulin/genetics
7.
Br J Cancer ; 120(2): 196-206, 2019 01.
Article in English | MEDLINE | ID: mdl-30498230

ABSTRACT

BACKGROUND: There are phase 3 clinical trials underway evaluating anti-PD-L1 antibodies as adjuvant (postoperative) monotherapies for resectable renal cell carcinoma (RCC) and triple-negative breast cancer (TNBC); in combination with antiangiogenic VEGF/VEGFR2 inhibitors (e.g., bevacizumab and sunitinib) for metastatic RCC; and in combination with chemotherapeutics as neoadjuvant (preoperative) therapies for resectable TNBC. METHODS: This study investigated these and similar clinically relevant drug combinations in highly translational preclinical models of micro- and macro-metastatic disease that spontaneously develop after surgical resection of primary kidney or breast tumours derived from orthotopic implantation of murine cancer cell lines (RENCAluc or EMT-6/CDDP, respectively). RESULTS: In the RENCAluc model, adjuvant sunitinib plus anti-PD-L1 improved overall survival compared to either drug alone, while the same combination was ineffective as early therapy for unresected primary tumours or late-stage therapy for advanced metastatic disease. In the EMT-6/CDDP model, anti-PD-L1 was highly effective as an adjuvant monotherapy, while its combination with paclitaxel chemotherapy (with or without anti-VEGF) was most effective as a neoadjuvant therapy. CONCLUSIONS: Our preclinical data suggest that anti-PD-L1 plus sunitinib may warrant further investigation as an adjuvant therapy for RCC, while anti-PD-L1 may be improved by combining with chemotherapy in the neoadjuvant but not the adjuvant setting of treating breast cancer.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Kidney Neoplasms/therapy , Mammary Neoplasms, Animal/therapy , Neovascularization, Pathologic/therapy , Animals , B7-H1 Antigen/immunology , Bevacizumab/administration & dosage , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/surgery , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/surgery , Mice , Neoadjuvant Therapy/methods , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Paclitaxel/administration & dosage , Sunitinib/administration & dosage
8.
Nat Rev Clin Oncol ; 15(5): 310-324, 2018 05.
Article in English | MEDLINE | ID: mdl-29434333

ABSTRACT

Immunotherapies have revolutionized medical oncology following the remarkable and, in some cases, unprecedented outcomes observed in certain groups of patients with cancer. Combination with other therapeutic modalities, including anti-angiogenic agents, is one of the many strategies currently under investigation to improve the response rates and duration of immunotherapies. Such a strategy might seem counterintuitive given that anti-angiogenic agents can increase tumour hypoxia and reduce the number of blood vessels within tumours. Herein, we review the additional effects mediated by drugs targeting VEGF-dependent signalling and other pathways, such as those mediated by angiopoietin 2 or HGF, which might increase the efficacy of immunotherapies. In addition, we discuss the seldom considered possibility that immunotherapies, and immune-checkpoint inhibitors in particular, might increase the efficacy of anti-angiogenic or other types of antivascular therapies and/or promote changes in the tumour vasculature. In short, we propose that interactions between both therapeutic modalities could be considered a 'two-way street'.


Subject(s)
Immunotherapy/trends , Molecular Targeted Therapy , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/therapeutic use , Angiopoietin-2/antagonists & inhibitors , Angiopoietin-2/immunology , Hepatocyte Growth Factor/antagonists & inhibitors , Hepatocyte Growth Factor/immunology , Humans , Immunologic Factors , Neoplasms/immunology , Neovascularization, Pathologic/immunology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology
9.
Cancer Cell ; 31(4): 469-471, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28399405

ABSTRACT

In this issue of Cancer Cell, Seaman et al. demonstrate that antibody drug conjugates (ADCs) against CD276 expressed by tumor cells and tumor vasculature have promising anti-tumor activity while showing little toxicity. Importantly, these agents have the potential to target both angiogenic vessels and non-angiogenic vessels co-opted by tumor cells.


Subject(s)
Antibodies , Cell Line, Tumor , Humans , Immunoconjugates
10.
Clin Exp Metastasis ; 33(2): 197-210, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26620208

ABSTRACT

Angiogenesis is a major requirement for tumour formation and development. Anti-angiogenic treatments aim to starve the tumour of nutrients and oxygen and also guard against metastasis. The main anti-angiogenic agents to date have focused on blocking the pro-angiogenic vascular endothelial growth factors (VEGFs). While this approach has seen some success and has provided a proof of principle that such anti-angiogenic agents can be used as treatment, the overall outcome of VEGF blockade has been somewhat disappointing. There is a current need for new strategies in inhibiting tumour angiogenesis; this article will review current and historical examples in blocking various membrane receptors and components of the extracellular matrix important in angiogenesis. Targeting these newly discovered pro-angiogenic proteins could provide novel strategies for cancer therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Animals , Humans , Vascular Endothelial Growth Factor A/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...