Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Br J Cancer ; 120(2): 196-206, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30498230

RESUMO

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.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Neoplasias Renais/terapia , Neoplasias Mamárias Animais/terapia , Neovascularização Patológica/terapia , Animais , Antígeno B7-H1/imunologia , Bevacizumab/administração & dosagem , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Imunoterapia/métodos , Neoplasias Renais/imunologia , Neoplasias Renais/patologia , Neoplasias Renais/cirurgia , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/cirurgia , Camundongos , Terapia Neoadjuvante/métodos , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Paclitaxel/administração & dosagem , Sunitinibe/administração & dosagem
2.
FASEB J ; 30(6): 2311-23, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26939791

RESUMO

C-type lectin family 14, member A (CLEC14A), is a single-pass transmembrane glycoprotein that is overexpressed in tumor endothelial cells, and it promotes sprouting angiogenesis and modulates endothelial function via interactions with extracellular matrix proteins. Here, we show that CLEC14A is cleaved by rhomboid-like protein 2 (RHBDL2), one of 3 catalytic mammalian rhomboid-like (RHBDL) proteases, but that it is not cleaved by RHBDL1 or -3. Site-directed mutagenesis identified the precise site at which RHBDL2 cleaves CLEC14A, and targeted, small interfering RNAs that knockdown endogenous CLEC14A and RHBDL2 in human endothelial cells validated the specificity of CLEC14A shedding by RHBDL2. Loss of endogenous cleaved CLEC14A increased endothelial migration 2-fold, whereas that addition of recombinant cleaved CLEC14A inhibited the sprouting of human and murine endothelial cells 3-fold in several in vitro models. We assessed the in vivo role of cleaved CLEC14A in angiogenesis by using the rodent subcutaneous sponge implant model, and we found that CLEC14A protein inhibited vascular density by >50%. Finally, we show that cleaved CLEC14A binds to sprouting endothelial tip cells. Our data show that the ectodomain of CLEC14A regulates sprouting angiogenesis and suggests a role for RHBDL2 in endothelial function.-Noy, P. J., Swain, R. K., Khan, K., Lodhia, P., Bicknell, R. Sprouting angiogenesis is regulated by shedding of the C-type lectin family 14, member A (CLEC14A) ectodomain, catalyzed by rhomboid-like 2 protein (RHBDL2).


Assuntos
Moléculas de Adesão Celular/metabolismo , Endopeptidases/metabolismo , Células Endoteliais/fisiologia , Lectinas Tipo C/metabolismo , Proteínas de Membrana/metabolismo , Neovascularização Fisiológica/fisiologia , Serina Proteases/metabolismo , Sequência de Aminoácidos , Animais , Fenômenos Biomecânicos , Moléculas de Adesão Celular/genética , Movimento Celular/fisiologia , Endopeptidases/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Lectinas Tipo C/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Domínios Proteicos , Serina Endopeptidases , Serina Proteases/genética
3.
Nat Rev Clin Oncol ; 21(6): 468-482, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38600370

RESUMO

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.


Assuntos
Inibidores da Angiogênese , Inibidores de Checkpoint Imunológico , Neoplasias , Humanos , Inibidores da Angiogênese/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ensaios Clínicos Fase III como Assunto , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
4.
J Biol Chem ; 287(47): 39753-65, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23035126

RESUMO

A disintegrin and metalloprotease 10 (ADAM10) is a ubiquitous transmembrane metalloprotease that cleaves the extracellular regions from over 40 different transmembrane target proteins, including Notch and amyloid precursor protein. ADAM10 is essential for embryonic development and is also important in inflammation, cancer, and Alzheimer disease. However, ADAM10 regulation remains poorly understood. ADAM10 is compartmentalized into membrane microdomains formed by tetraspanins, which are a superfamily of 33 transmembrane proteins in humans that regulate clustering and trafficking of certain other transmembrane "partner" proteins. This is achieved by specific tetraspanin-partner interactions, but it is not clear which tetraspanins specifically interact with ADAM10. The aims of this study were to identify which tetraspanins interact with ADAM10 and how they regulate this metalloprotease. Co-immunoprecipitation identified specific ADAM10 interactions with Tspan5, Tspan10, Tspan14, Tspan15, Tspan17, and Tspan33/Penumbra. These are members of the largely unstudied TspanC8 subgroup of tetraspanins, all six of which promoted ADAM10 maturation. Different cell types express distinct repertoires of TspanC8 tetraspanins. Human umbilical vein endothelial cells express relatively high levels of Tspan14, the knockdown of which reduced ADAM10 surface expression and activity. Mouse erythrocytes express predominantly Tspan33, and ADAM10 expression was substantially reduced in the absence of this tetraspanin. In contrast, ADAM10 expression was normal on Tspan33-deficient mouse platelets in which Tspan14 is the major TspanC8 tetraspanin. These results define TspanC8 tetraspanins as essential regulators of ADAM10 maturation and trafficking to the cell surface. This finding has therapeutic implications because focusing on specific TspanC8-ADAM10 complexes may allow cell type- and/or substrate-specific ADAM10 targeting.


Assuntos
Proteínas ADAM/biossíntese , Secretases da Proteína Precursora do Amiloide/biossíntese , Regulação Enzimológica da Expressão Gênica/fisiologia , Microdomínios da Membrana/enzimologia , Proteínas de Membrana/biossíntese , Tetraspaninas/metabolismo , Proteínas ADAM/genética , Proteína ADAM10 , Doença de Alzheimer/enzimologia , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Plaquetas/citologia , Plaquetas/enzimologia , Linhagem Celular , Eritrócitos/citologia , Eritrócitos/enzimologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/enzimologia , Inflamação/genética , Inflamação/patologia , Microdomínios da Membrana/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Ligação Proteica , Transporte Proteico/fisiologia , Tetraspaninas/genética
5.
Sci Immunol ; 7(67): eabm6388, 2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35030032

RESUMO

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.


Assuntos
Células Endoteliais/imunologia , Neoplasias/imunologia , Neovascularização Patológica/imunologia , Animais , Humanos , Imunoterapia , Neoplasias/terapia , Neovascularização Patológica/terapia
6.
Biochem Soc Trans ; 39(6): 1667-73, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22103505

RESUMO

Tetraspanins function as organizers of the cell surface by recruiting specific partner proteins into tetraspanin-enriched microdomains, which regulate processes such as cell adhesion, signalling and intracellular trafficking. Endothelial cells appear to express at least 23 of the 33 human tetraspanins, and a number of recent studies have demonstrated their importance in endothelial cell biology. Tetraspanin CD151 is essential for pathological angiogenesis, which may in part be due to regulation of its main partner proteins, the laminin-binding integrins α3ß1, α6ß1 and α6ß4. CD9 and CD151 are essential for leucocyte recruitment during an inflammatory response, through the formation of pre-assembled nano-platforms containing the adhesion molecules ICAM-1 (intercellular adhesion molecule 1) and VCAM-1 (vascular cell adhesion molecule 1), which ultimately coalesce to form docking structures around captured leucocytes. Tetraspanin CD63 also facilitates leucocyte capture by promoting clustering of the adhesion molecule P-selectin. Finally, Tspan12 is required for blood vessel development in the eye, through regulation of Norrin-induced Frizzled-4 signalling, such that Tspan12 mutations can lead to human disease. Future studies on these and other endothelial tetraspanins are likely to provide further novel insights into angiogenesis and inflammation.


Assuntos
Células Endoteliais/metabolismo , Microdomínios da Membrana/metabolismo , Tetraspaninas/metabolismo , Animais , Vasos Sanguíneos/crescimento & desenvolvimento , Vasos Sanguíneos/metabolismo , Adesão Celular , Células Endoteliais/citologia , Humanos , Neovascularização Fisiológica , Tetraspaninas/química
7.
EMBO Mol Med ; 13(7): e08253, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34125494

RESUMO

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.


Assuntos
Angiopoietina-2 , Neoplasias , Inibidores da Angiogênese/uso terapêutico , Angiopoietina-1 , Humanos , Imunoterapia , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Receptor TIE-2
8.
EMBO Mol Med ; 12(6): e12355, 2020 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-32406209

RESUMO

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.


Assuntos
Neoplasias , Anticorpos , Humanos , Metástase Neoplásica , Neoplasias/tratamento farmacológico
9.
NPJ Breast Cancer ; 6: 29, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32704531

RESUMO

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.

10.
FEBS J ; 286(17): 3299-3332, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31287944

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Endotélio Vascular/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Complemento/metabolismo , Trombomodulina/metabolismo , Animais , Antígenos CD/genética , Antígenos de Neoplasias/genética , Moléculas de Adesão Celular/genética , Humanos , Lectinas Tipo C/genética , Glicoproteínas de Membrana/genética , Neoplasias/metabolismo , Neovascularização Fisiológica , Receptores de Complemento/genética , Trombomodulina/genética
11.
Nat Rev Clin Oncol ; 15(5): 310-324, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29434333

RESUMO

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'.


Assuntos
Imunoterapia/tendências , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Angiopoietina-2/antagonistas & inibidores , Angiopoietina-2/imunologia , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/imunologia , Humanos , Fatores Imunológicos , Neoplasias/imunologia , Neovascularização Patológica/imunologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/imunologia
12.
Cancer Cell ; 31(4): 469-471, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28399405

RESUMO

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.


Assuntos
Anticorpos , Linhagem Celular Tumoral , Humanos , Imunoconjugados
13.
Sci Signal ; 10(481)2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28559451

RESUMO

NKG2D (natural killer group 2, member D) is an activating receptor found on the surface of immune cells, including natural killer (NK) cells, which regulates innate and adaptive immunity through recognition of the stress-induced ligands ULBP1 (UL16 binding protein 1) to ULBP6 and MICA/B. Similar to class I human leukocyte antigen (HLA), these NKG2D ligands have a major histocompatibility complex-like fold and exhibit pronounced polymorphism, which influences human disease susceptibility. However, whereas class I HLA polymorphisms occur predominantly in the α1α2 groove and affect antigen binding, the effects of most NKG2D ligand polymorphisms are unclear. We studied the molecular and functional consequences of the two major alleles of ULBP6, the most polymorphic ULBP gene, which are associated with autoimmunity and relapse after stem cell transplantation. Surface plasmon resonance and crystallography studies revealed that the arginine-to-leucine polymorphism within ULBP0602 affected the NKG2D-ULBP6 interaction by generating an energetic hotspot. This resulted in an NKG2D-ULBP0602 affinity of 15.5 nM, which is 10- to 1000-fold greater than the affinities of other ULBP-NKG2D interactions and limited NKG2D-mediated activation. In addition, soluble ULBP0602 exhibited high-affinity competitive binding for NKG2D and partially suppressed NKG2D-mediated activation of NK cells by other NKG2D ligands. These effects resulted in a decrease in a range of NKG2D-mediated effector functions. Our results reveal that ULBP polymorphisms affect the strength of human lymphocyte responses to cellular stress signals and may offer opportunities for therapeutic intervention.


Assuntos
Neoplasias Hematológicas/patologia , Células Matadoras Naturais/patologia , Proteínas de Membrana/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Polimorfismo Genético , Linfócitos T/patologia , Ligação Competitiva , Membrana Celular/metabolismo , Células Cultivadas , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ligantes , Proteínas de Membrana/química , Proteínas de Membrana/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/química , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Conformação Proteica , Linfócitos T/imunologia , Linfócitos T/metabolismo
14.
Clin Exp Metastasis ; 33(2): 197-210, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26620208

RESUMO

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.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Animais , Humanos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA