scholarly journals Pools of programmed death-ligand within the oral cavity tumor microenvironment: Variable alteration by targeted therapies

Head & Neck ◽  
2016 ◽  
Vol 38 (8) ◽  
pp. 1176-1186 ◽  
Author(s):  
Sujay Shah ◽  
Andria Caruso ◽  
Harrison Cash ◽  
Carter Van Waes ◽  
Clint T. Allen

2019 ◽  
Vol 5 (1) ◽  
Author(s):  
David B. Page ◽  
Harry Bear ◽  
Sangeetha Prabhakaran ◽  
Margaret E. Gatti-Mays ◽  
Alexandra Thomas ◽  
...  

Abstract Antibodies blocking programmed death 1 (anti-PD-1) or its ligand (anti-PD-L1) are associated with modest response rates as monotherapy in metastatic breast cancer, but are generally well tolerated and capable of generating dramatic and durable benefit in a minority of patients. Anti-PD-1/L1 antibodies are also safe when administered in combination with a variety of systemic therapies (chemotherapy, targeted therapies), as well as with radiotherapy. We summarize preclinical, translational, and preliminary clinical data in support of combination approaches with anti-PD-1/L1 in metastatic breast cancer, focusing on potential mechanisms of synergy, and considerations for clinical practice and future investigation.



2018 ◽  
Vol 52 (1) ◽  
pp. 119-120 ◽  
Author(s):  
W. Ventura ◽  
R. H. Novoa ◽  
C. I. Lazarte‐Rantes ◽  
W. Castillo‐Urquiaga ◽  
M. Zárate‐Girao ◽  
...  


2019 ◽  
Vol 79 (16) ◽  
pp. 4149-4159 ◽  
Author(s):  
Renee Clift ◽  
Jennifer Souratha ◽  
Sheryl A. Garrovillo ◽  
Susan Zimmerman ◽  
Barbara Blouw


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2941-2941
Author(s):  
Jianfei Qian ◽  
Yuhuan Zheng ◽  
Lijuan Wang ◽  
Sungyoul Hong ◽  
Haiyan Li ◽  
...  

Abstract Abstract 2941 Current approaches for vaccination and immunotherapy are often capable of eliciting strong T-cell responses against tumor antigens. However, negative regulatory mechanisms within the tumor microenvironment inhibit antitumor T-cell function, leading to evasion from immune attack. One inhibitory mechanism is upregulation of programmed death-ligand 1 (PD-L1/B7H1) expressed on tumor or stromal cells, which binds to programmed death-1 (PD-1) on activated T cells. PD-1 and B7H1 engagement results in diminished antitumor T-cell responses and correlates with poor outcome in murine and human cancers. To cure mice with large tumor burdens by administering tumor vaccines, we hypothesized that it is necessary not only to enhance the immunogenicity of the vaccines, but also to target the suppressive tumor microenvironment and break immune suppression on effector cells. Therefore, we explored combinational treatments in a myeloma setting by using vaccination and anti-B7H1 blocking mAbs to enhance clinical efficacy of cancer vaccines. To establish mouse tumor models for evaluation of tumor-associated B7H1 in the inhibition of T-cell immunity, first, we examined the expression of B7H1 in four murine myeloma cell lines that originated from Balb/c mice. Results showed that all the myeloma cells expressed a negative T-cell costimulatory molecule B7H1, and blocking surface B7H1 by using a specific mAb (M5H1) enabled tumor-specific CTLs from vaccinated mice to be more efficient at lysing the tumor cells. Thus these results suggest that B7H1 on tumor cells and PD-1 on T cells may form a B7H1/PD-1 molecular shield to prevent lysis by T cells and actively inhibit the cytolytic function of tumor-specific CTLs by expressing B7H1. Next, we evaluated the therapeutic efficacy of DKK1-DNA vaccine in mice with established myeloma. In these experiments, DKK1-DNA vaccine plus CpG was used as a standard vaccine. Anti-B7H1 (M5H1) mAb was used to block negative T-cell signaling in mice receiving DKK1-DNA vaccination. Balb/c mice were first inoculated subcutaneously with the myeloma cells (1 million cells per mouse). Ten days after tumor inoculation, mice were immunized with three injections of the vaccines on days 10, 14, and 17 after tumor inoculation. Mice were also injected intraperitoneally with B7H1 (200 μg per mouse per treatment) on days 1, 4, 7, and 10 following the first vaccination. Mice receiving injections of PBS, isotype IgG (200 μg per mouse per treatment), or CpG- (50 μg per mouse per treatment) alone were used as controls. Results show that mice receiving DKK1-DNA vaccine (plus CpG) together with B7H1 induced more robust tumor growth inhibition (P < 0.01, compared with PBS control mice). The survival of DKK1-CpG vaccine- and vaccine plus B7H1 antibody-treated mice were 60% and 80%, respectively. Thus, our data showed that B7H1 blocking mAbs can further enhance the therapeutic efficacy of the vaccine. Finally, we investigated the mechanism of therapeutic efficacy of vaccine enhanced by anti-B7H1 mAbs. Our results showed that the percentages of IL-10-secreting or Foxp3+ Tregs in the spleens and tumors are significantly decreased in mice vaccinated with DKK1 DNA plus CpG in combination with anti-B7H1 antibodies (P < 0.01, compared with mice receiving DKK1 DNA alone or vaccine plus CpG). These results indicate that myeloma cells are able to induce or recruit regulatory T cells, and the beneficial effects of anti-B7H1 mAbs were derived from their ability to reduce the numbers of Tregs in tumor-bearing mice. Taken together, these results support the concept that B7H1/PD-1 forms a molecular shield to prevent destruction by CTLs and implicate new approaches for immunotherapy of human cancers by B7H1 blocking mAbs. Disclosures: No relevant conflicts of interest to declare.



2014 ◽  
Vol 2014 ◽  
pp. 1-8 ◽  
Author(s):  
Pilar de la Puente ◽  
Barbara Muz ◽  
Feda Azab ◽  
Micah Luderer ◽  
Abdel Kareem Azab

Multiple myeloma (MM) is a hematological malignancy that remains incurable because most patients will eventually relapse or become refractory to the treatments. Although the treatments have improved, the major problem in MM is the resistance to therapy. Novel agents are currently in development for the treatment of relapsed/refractory MM, including immunomodulatory drugs, proteasome inhibitors, monoclonal antibodies, cell signaling targeted therapies, and strategies targeting the tumor microenvironment. We have previously reviewed in detail the contemporary immunomodulatory drugs, proteasome inhibitors, and monoclonal antibodies therapies for MM. Therefore, in this review, we focused on the role of molecular targeted therapies in the treatment of relapsed/refractory multiple myeloma, including cell signaling targeted therapies (HDAC, PI3K/AKT/mTOR, p38 MAPK, Hsp90, Wnt, Notch, Hedgehog, and cell cycle) and strategies targeting the tumor microenvironment (hypoxia, angiogenesis, integrins, CD44, CXCR4, and selectins). Although these novel agents have improved the therapeutic outcomes for MM patients, further development of new therapeutic agents is warranted.



Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 777-777
Author(s):  
Fengdong Cheng ◽  
Yuan Ren ◽  
Jie Chen ◽  
Zi Wang ◽  
Tao Li ◽  
...  

Abstract Remarkable clinical efficacy and durable responses to antibodies that block the programmed death-1 (PD-1)-programmed death-ligand 1 (PD-L1) pathway have been observed in patients with multiple cancers, including classical Hodgkin lymphomas (cHL). However, the responses in the majority of Non-Hodgkin lymphoma patients, including mantle cell lymphoma (MCL), treated with anti- PD1/PDL1 antibodies have been modest to date. It has been postulated that the immune suppressive nature of the tumor microenvironment (TME) may play a role in limiting the efficacy of checkpoint blockade strategies. As such, identification of critical molecules in TME required for driving response and resistance is key to improve lymphoma immunotherapy. We have therefore generated in vivo and ex-vivo MCL lymphoma-stroma co-culture models and capitalize this model with primary human MCL cells as well. First, we found that co-injection of murine Fc-muMCL1 cells with stromal cells significantly promote lymphoma growthas compared to Fc-muMCL1 cells injected alone. This aggressive growth was associated with less tumor infiltrating cytotoxic T-cells in the TME. Second, to identify the tolerogenic mechanism(s) that drive immunosuppression in TME, we co-cultured MCL cells with stroma cells ex-vivo and found an increased translation and transcription of PD-L1 via upregulation of c-Myc. Furthermore, co-culture of patient primary lymphoma cells with stromal cell dramatically increases PD-L1 expression in both stromal cells and lymphoma cells. Tumor infiltrating T cells also induce PD-L1 expression in stromal cells. Of note, just by knocking down c-Myc in stromal cell we were able to block co-culture-induced PD-L1 expression, highlighting a critical role for c-Myc in driving this tolerogenic process in the TME. In lieu of the above findings, next we treated murine MCL in vitro with a bromodomain inhibitor (JQ1) and observed a significant decrease in c-Myc/PD-L1 expression which was associated with increased immunogenicity of malignant B-cells leading to a better T-cell activation. More importantly, treatment of MCL-bearing mice with a combination of a bromodomain inhibitor with anti-PD1 antibody resulted in enhanced inhibition of MCL growth, increased effector memory T cells and improved function of tumor infiltrating T cells in vivo. No such effects were observed in MCL-bearing mice treated with either agent alone. Taken together, we have identified the c-Myc/PD-L1 axis in stromal cells that by creating a tolerogenic/immunosuppressive TME imposes a significant barrier to the efficacy of checkpoint blockade therapy in lymphomas. This barrier seems not to be unsurmountable since the addition of a bromodomain inhibitor augmented the efficacy of checkpoint blockade by inducing a more immunogenic TME in MCL. Disclosures No relevant conflicts of interest to declare.



2021 ◽  
Vol 2 ◽  
Author(s):  
Alex Dobriyan ◽  
Iris Gluck ◽  
Eran Alon ◽  
Iris Barshack ◽  
Ran Yahalom ◽  
...  

Background: The tumor microenvironment (TME) of oral squamous cell carcinoma (OSCC) is associated with immune suppression, one of the pathways being the programmed death receptor 1 (PD-1) and its ligands (PD-L1/PD-L2). Checkpoint inhibitors of PD-1/PD-L1, like pembrolizumab, have been recently approved for treatment of OSCC. We described the histologic findings in OSCC following neoadjuvant pembrolizumab, including identification of immune-related cell populations and cancer-associated fibroblasts (CAFs).Materials and Methods: Patients with OSCC clinical stages 3 and 4 and a combined PD-L1 score &gt;1 were randomized either to the standard oncologic protocol or to the pembrolizumab arm of MK-3475-689 study for Head and Neck, Lip, and Oral Cavity. The latter were given two standard doses of 200 mg of pembrolizumab, 3 weeks apart, and then underwent surgical oncologic procedure according to the initial stage. Sections from the resection specimens were analyzed for pathological response to pembrolizumab. Various populations of immune-related cells within the tumor microenvironment were characterized by immunohistochemistry, as were the CAFs.Results: Three patients who were randomized to the pembrolizumab study were described. One patient presented with a tongue SCC, the other two had SCC of the mandibular ridge with bony involvement. Only the patient with tongue SCC showed clinical complete response. Microscopically, the tumor was replaced by a granulomatous type of inflammation. Immunohistochemical stains revealed massive T cell rich (CD3+) infiltrate, with approximately equal amounts of CD4+ and CD8+ cells, numerous macrophages of CD68+ and CD163+ phenotypes; no CAFs were identified. The other two patients were regarded as non-responders as at least 50% of the tumor was viable. The tumor microenvironment of these tumors was generally associated with a lesser extent of inflammatory response compared to the tongue tumor, a variable CD4+/CD8+ ratio and presence of CAFs. Neither T regulatory cells (FOXP3+) nor natural killer cells (CD56+, CD57+) were identified in any of the cases.Conclusion: We showed that characterizing the specific populations of immune-related cells and CAFs after treatment with pembrolizumab, may add to our understanding of the tumor-TME interactions in this setting. These findings should be investigated in future studies on a larger number of patients.



2021 ◽  
Vol 9 ◽  
pp. 2050313X2110658
Author(s):  
McKenna Hawthorne ◽  
Tristan Tham ◽  
Brent Ponder ◽  
Alexandros Georgolios

Reactive hyperplasia is a phenomenon responsible for exophytic lesions in the oral cavity, and may appear to be suspicious, especially in patients who have a significant history of malignancy. Here, we present a case of reactive hyperplasia mimicking recurrence in a patient who was previously treated for tonsillar carcinoma. Physicians who commonly see patients with oral lesions, particularly oral surgeons and otolaryngologists, should be cognizant of the unusual presentation of these lesions as they may mimic the physical characteristics of recurrence.



2021 ◽  
Vol 11 ◽  
Author(s):  
Hye-Youn Son ◽  
Hwan-Kyu Jeong

Extensive interest in cancer immunotherapy is reported according to the clinical importance of CTLA-4 and (PD-1/PD-L1) [programmed death (PD) and programmed death-ligand (PD-L1)] in immune checkpoint therapies. AXL is a receptor tyrosine kinase expressed in different types of cancer and in relation to resistance against various anticancer therapeutics due to poor clinical prognosis. AXL and its ligand, i.e., growth arrest-specific 6 (GAS6) proteins, are expressed on many cancer cells, and the GAS6/AXL pathway is reported to promote cancer cell proliferation, survival, migration, invasion, angiogenesis, and immune evasion. AXL is an attractive and novel therapeutic target for impairing tumor progression from immune cell contracts in the tumor microenvironment. The GAS6/AXL pathway is also of interest immunologically because it targets fewer antitumor immune responses. In effect, several targeted therapies are selective and nonselective for AXL, which are in preclinical and clinical development in multiple cancer types. Therefore, this review focuses on the role of the GAS6/AXL signaling pathway in triggering the immunosuppressive tumor microenvironment as immune evasion. This includes regulating its composition and activating T-cell exclusion with the immune-suppressive activity of regulatory T cells, which is related to one of the hallmarks of cancer survival. Finally, this article discusses the GAS6/AXL signaling pathway in the context of several immune responses such as NK cell activation, apoptosis, and tumor-specific immunity, especially PD-1/PDL-1 signaling.



Sign in / Sign up

Export Citation Format

Share Document