2005
DOI: 10.1158/0008-5472.1089.65.3
|View full text |Cite
|
Sign up to set email alerts
|

Blockade of B7-H1 and PD-1 by Monoclonal Antibodies Potentiates Cancer Therapeutic Immunity

Abstract: Contemporary approaches for vaccination and immunotherapy are often capable of eliciting strong T-cell responses against tumor antigens. However, such responses are not parallel to clinical tumor regression. The development of evasion mechanisms within tumor microenvironment may be responsible for poor therapeutic responses. We report here that constitutive or inducible expression of B7-H1, a B7 family molecule widely expressed by cancers, confers resistance to therapeutic anti-CD137 antibody in mice with esta… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
4
1

Citation Types

2
74
0
3

Year Published

2016
2016
2022
2022

Publication Types

Select...
5
4

Relationship

0
9

Authors

Journals

citations
Cited by 844 publications
(81 citation statements)
references
References 35 publications
2
74
0
3
Order By: Relevance
“…The immune system plays an important role in cancer initiation and progression . Various mechanisms can be developed by cancer cells to escape from immune surveillance, , such as immune checkpoints, which refer to inhibitory receptors expressed on immune effector cells or immune inhibitory ligands on tumor cells. , Various immune checkpoint molecules have been discovered, such as programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and others. Blockade of PD-1 or CTLA-4 using monoclonal antibodies (mAbs) has led to many exciting preclinical and clinical results, as shown by the enhanced and sustained endogenous antitumor immune responses, resulting in durable tumor control. , Despite the successes in some cancer patients, the overall clinical efficacy of the checkpoint blockade is still limited due to the fact that other suppressive mechanisms are also involved in immune escape, such as increased expression of indoleamine 2,3-dioxygenase (IDO). , Another reason is that the efficacy of this treatment is closely associated with pre-existing antitumor immune responses. , Chemotherapy drugs can trigger immunogenic cell death and initiate antitumor immune responses. , The checkpoint blockade can then further enhance and sustain such antitumor immunity. Therefore, one ideal strategy will be to induce immunogenic tumor cell death while simultaneously blocking multiple tumor immunosuppressive pathways.…”
Section: Introductionmentioning
confidence: 99%
See 1 more Smart Citation
“…The immune system plays an important role in cancer initiation and progression . Various mechanisms can be developed by cancer cells to escape from immune surveillance, , such as immune checkpoints, which refer to inhibitory receptors expressed on immune effector cells or immune inhibitory ligands on tumor cells. , Various immune checkpoint molecules have been discovered, such as programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), and others. Blockade of PD-1 or CTLA-4 using monoclonal antibodies (mAbs) has led to many exciting preclinical and clinical results, as shown by the enhanced and sustained endogenous antitumor immune responses, resulting in durable tumor control. , Despite the successes in some cancer patients, the overall clinical efficacy of the checkpoint blockade is still limited due to the fact that other suppressive mechanisms are also involved in immune escape, such as increased expression of indoleamine 2,3-dioxygenase (IDO). , Another reason is that the efficacy of this treatment is closely associated with pre-existing antitumor immune responses. , Chemotherapy drugs can trigger immunogenic cell death and initiate antitumor immune responses. , The checkpoint blockade can then further enhance and sustain such antitumor immunity. Therefore, one ideal strategy will be to induce immunogenic tumor cell death while simultaneously blocking multiple tumor immunosuppressive pathways.…”
Section: Introductionmentioning
confidence: 99%
“…5−7 Blockade of PD-1 or CTLA-4 using monoclonal antibodies (mAbs) has led to many exciting preclinical and clinical results, as shown by the enhanced and sustained endogenous antitumor immune responses, resulting in durable tumor control. 8,9 Despite the successes in some cancer patients, the overall clinical efficacy of the checkpoint blockade is still limited due to the fact that other suppressive mechanisms are also involved in immune escape, such as increased expression of indoleamine 2,3-dioxygenase (IDO). 10,11 Another reason is that the efficacy of this treatment is closely associated with preexisting antitumor immune responses.…”
Section: ■ Introductionmentioning
confidence: 99%
“…29 We last examined how well the soluble αPD-1-iTEP fusion and αPD-1 NP inhibit the binding of PD-L1 to the PD-1-positive cells, the working mechanism of PD-1 immune checkpoint therapy. 30,31 To accomplish the examination, we designed and employed a PD-L1 binding inhibition assay. Specifically, we took advantage of the facts that PD-L1-human Fc, a fusion protein of mouse PD-L1 fusion and human IgG Fc, bound with EL4 cells, and that the bound PD-L1-human Fc can be detected by an anti-human Fc antibody.…”
Section: Molecular Pharmaceuticsmentioning
confidence: 99%
“…22). Pembrolizumab and nivolumab are immune checkpoint inhibitors (ICI) that block programmed death-1 (PD-1) and reinvigorate exhausted antitumor CD8 þ T cells within the tumor microenvironment (23)(24)(25). Pembrolizumab and nivolumab produce durable clinical responses and are approved in multiple cancers (26)(27)(28)(29)(30)(31)(32).…”
Section: Introductionmentioning
confidence: 99%