Clinical Trials Investigating Immune Checkpoint Blockade in Glioblastoma

Author(s):  
Russell Maxwell ◽  
Christopher M. Jackson ◽  
Michael Lim
Vaccines ◽  
2022 ◽  
Vol 10 (1) ◽  
pp. 70
Author(s):  
Takumi Kumai ◽  
Hidekiyo Yamaki ◽  
Michihisa Kono ◽  
Ryusuke Hayashi ◽  
Risa Wakisaka ◽  
...  

The success of the immune checkpoint blockade has provided a proof of concept that immune cells are capable of attacking tumors in the clinic. However, clinical benefit is only observed in less than 20% of the patients due to the non-specific activation of immune cells by the immune checkpoint blockade. Developing tumor-specific immune responses is a challenging task that can be achieved by targeting tumor antigens to generate tumor-specific T-cell responses. The recent advancements in peptide-based immunotherapy have encouraged clinicians and patients who are struggling with cancer that is otherwise non-treatable with current therapeutics. By selecting appropriate epitopes from tumor antigens with suitable adjuvants, peptides can elicit robust antitumor responses in both mice and humans. Although recent experimental data and clinical trials suggest the potency of tumor reduction by peptide-based vaccines, earlier clinical trials based on the inadequate hypothesis have misled that peptide vaccines are not efficient in eliminating tumor cells. In this review, we highlighted the recent evidence that supports the rationale of peptide-based antitumor vaccines. We also discussed the strategies to select the optimal epitope for vaccines and the mechanism of how adjuvants increase the efficacy of this promising approach to treat cancer.


2014 ◽  
Vol 21 (2) ◽  
pp. 371-381 ◽  
Author(s):  
Mabel Ryder ◽  
Margaret Callahan ◽  
Michael A Postow ◽  
Jedd Wolchok ◽  
James A Fagin

Novel immune checkpoint blockade with ipilimumab, an antibody blocking the cytotoxic T-lymphocyte antigen 4 (CTLA4), is revolutionizing cancer therapy. However, ipilimumab induces symptomatic, sometimes severe, endocrine immune-related adverse events (irAEs) that are inconsistently recognized and reported. The objective of this review was to comprehensively characterize the incidence, presentation, and management of endocrinopathies following ipilimumab therapy in a single center that is highly specialized in immune checkpoint blockade. We carried out a retrospective analysis of endocrine irAEs in melanoma patients receiving ipilimumab therapy in clinical trials between 2007 and 2013. A total of 256 patients were included in this analysis. We reviewed pituitary-, thyroid-, and adrenal-related hormone test results, as well as radiographic studies and the clinical histories of patients, to identify and characterize cases of hypophysitis, hypothyroidism, thyroiditis, and adrenal dysfunction. Following ipilimumab therapy, the overall incidence of hypophysitis was 8% and that of hypothyroidism/thyroiditis 6%. Primary adrenal dysfunction was rare. Therapy with a combination of ipilimumab and nivolumab, an anti-programmed cell death 1 (PDCD1, also called PD1) receptor antibody, was associated with a 22% incidence of either thyroiditis or hypothyroidism and a 9% incidence of hypophysitis. Symptomatic relief, in particular, for hypophysitis, was achieved in all patients with hormone replacement, although endogenous hormone secretion rarely recovered. In summary, we observed that CTLA4 blockade alone, and in particular in combination with PD1 blockade, is associated with an increased risk of symptomatic, sometimes severe, hypophysitis as well as thyroid dysfunction. Prompt initiation with hormone replacement reverses symptoms. Evaluation and reporting of endocrine irAEs in clinical trials should be done using standardized diagnostic criteria and terminology.


Author(s):  
Gary Middleton

Abstract In this first in a series of ‘Trials Watch’ articles we briefly review a highly selected set of clinical trials that are currently recruiting or about to open to recruitment in melanoma, the disease first transformed by the introduction of immune checkpoint blockade inhibitors (ICI). We place equal emphasis on phase I/II studies investigating the activity of biologically compelling novel immunotherapeutics, and on randomised trials of ICI with and without novel agents, as these latter studies optimise the standard of care use of ICI, and determine whether novel agents become part of the approved therapeutic armamentarium. We do not consider here combination therapy with other checkpoint antagonists or agonists besides combination of anti-PD-1/PD-L1 monoclonal antibodies (mAbs) with anti-CTLA4 mAbs, as these will be reviewed in a subsequent article in this series. A glossary of agents to be discussed is found at the end of this article.


2021 ◽  
Vol 11 ◽  
Author(s):  
Muhammet Ozer ◽  
Andrew George ◽  
Suleyman Yasin Goksu ◽  
Thomas J. George ◽  
Ilyas Sahin

The prevalence of primary liver cancer is rapidly rising all around the world. Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer. Unfortunately, the traditional treatment methods to cure HCC showed poor efficacy in patients who are not candidates for liver transplantation. Until recently, tyrosine kinase inhibitors (TKIs) were the front-line treatment for unresectable liver cancer. However, rapidly emerging new data has drastically changed the landscape of HCC treatment. The combination treatment of atezolizumab plus bevacizumab (immunotherapy plus anti-VEGF) was shown to provide superior outcomes and has become the new standard first-line treatment for unresectable or metastatic HCC. Currently, ongoing clinical trials with immune checkpoint blockade (ICB) have focused on assessing the benefit of antibodies against programmed cell death 1 (PD-1), programmed cell death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte- associated antigen 4 (CTLA-4) as monotherapies or combination therapies in patients with HCC. In this review, we briefly discuss the mechanisms underlying various novel immune checkpoint blockade therapies and combination modalities along with recent/ongoing clinical trials which may generate innovative new treatment approaches with potential new FDA approvals for HCC treatment in the near future.


2019 ◽  
Vol 19 (2) ◽  
pp. 222-228 ◽  
Author(s):  
Fan Zhongqi ◽  
Sun Xiaodong ◽  
Chen Yuguo ◽  
Lv Guoyue

Background: Hepatocellular Carcinoma (HCC) is one of the most common cancers with high mortality rate. The effects of most therapies are limited. The Immune Checkpoint Blockade (ICB) improves the prognosis in multiple malignancies. The application of immune checkpoint blockade to hepatocellular carcinoma patients has recently started. Early phase clinical trials have shown some benefits to cancer patients. Methods/Results: This review focuses on the immune system of liver and clinical trials of ICB. In particular, we analyze the mechanisms by which immune checkpoint blockade therapies can be used for the treatment of hepatocellular carcinoma patients, then examine the factors in cancer resistance to the therapies and finally suggest possible combination therapies for the treatment of hepatocellular carcinoma patients. Conclusion: ICB is a promising therapy for advanced HCC patients. Combined therapy exhibits a great potential to enhance ICB response in these patients. The better understanding of the factors influencing the sensitivity of ICB and more clinical trials will consolidate the efficiency and minimize the adverse effects of ICB.


2017 ◽  
Vol 35 (6_suppl) ◽  
pp. 356-356 ◽  
Author(s):  
Samuel Funt ◽  
Zhenyu Mu ◽  
Catharine Kline Cipolla ◽  
Brooke Elizabeth Kania ◽  
Junting Zheng ◽  
...  

356 Background: M-MDSCs promote tumor progression through complex mechanisms, including immunosuppression and the production of mediators of angiogenesis and invasion. M-MDSCs are associated with poor outcomes in a number of malignancies. We conducted an exploratory analysis enumerating M-MDSCs (Lin-CD14+/HLA-DRlow/-) in the blood of pts with mUC and assessed for assay variability and correlation with clinical outcomes. Methods: Whole blood was collected at a single time point for each pt and stabilized in Cyto-Chex tubes. M-MDSC% was calculated by flow cytometric analysis of HLA-DR expression on CD14+ monocytes using an MSKCC developed computational algorithm-based approach (Kitano et al. 2014). Individual samples were run in quadruplicate at MSKCC. The mean MDSC% was used in subsequent analyses and the replicate standard deviation (SD) was considered a reflection of intra-assay variability. Clinical variables were collected and pts followed for OS, which was calculated from time of sample collection. Results: 21 pts with mUC who progressed after prior chemotherapy were included. At the time of collection, pts were on clinical trials with immune checkpoint blockade (n=13) or targeted therapy (n=1), receiving salvage chemotherapy (n=2), or awaiting treatment on clinical trials with immune checkpoint blockade (n=4) or targeted therapy (n=1). The median follow up from the time of collection in surviving patients (n=15) was 11.1 months (range: 10.5-11.5). Median OS was not reached. Mean (SD) M-MDSC% was 29.2 (4.92). The range of replicate SD was 0.13-1.05. M-MDSC% was not higher in pts with visceral mets (p=0.109). Pts with above median M-MDSC% had worse OS (p=0.01; 6 patients died during follow up, all with above median M-MDSC%). Conclusions: In a heterogeneous cohort of pts with mUC, the enumeration of M-MDSCs in stabilized whole blood was feasible and demonstrated marked inter-patient but not intra-assay variability. Pts with higher M-MDSC% values had worse OS. Additional characterization of M-MDSCs in larger cohorts and in pts receiving immunotherapy is ongoing and may have important prognostic and therapeutic implications in mUC.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4416-4416
Author(s):  
Aidi Gu ◽  
Huaxian Ma ◽  
Xiaorui Zhang ◽  
Prerna Malaney ◽  
Miguel Gallardo ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) is often driven by aberrant activation of the B-cell receptor signaling pathway. We and others have shown in preclinical animal models as well as human clinical trials that the BTK inhibitor, ibrutinib, is effective in reducing CLL tumor burden and improving survival rates in both humans and mice. Preclinical studies also suggest that BTK inhibitors likely act not only through their direct effect on CLL tumor cells but also by alleviating immunosuppression in the tumor microenvironment through BTK inhibition in myeloid-derived suppressor cells. Additionally, we have previously demonstrated that CLL cells are susceptible to cytotoxic killing by T cells targeting the aberrantly expressed TCL1 oncoprotein (Weng et al. Blood 2012). Therefore, we hypothesized that the combination of BTK-pathway inhibition in conjunction with activation of antigen-specific T-cells by immune checkpoint blockade will be a synergistic therapeutic strategy. Here, we examined the effect of acalabrutinib (previously known as ACP-196), a selective BTK inhibitor with limited effect on other kinases, alone or in combination with immune checkpoint blockade in a mouse model of CLL. Eµ-TCL1 mice, which overexpress TCL1 in B-cells, were used as a model of CLL. We generated cohorts of Eµ-TCL1 mice and treated them with anti-PD-1 antibody (n = 18), acalabrutinib (n = 20), and acalabrutinib+anti-PD-1 antibody (n= 19). Treatment cohorts had a significant reduction in CD5+CD19+ tumor cells in peripheral blood and spleens as compared to vehicle-treated mice (n =16) (p <0.05). However, the most pronounced anti-tumor effects were observed in mice treated with acalabrutinib alone. In fact, MRI imaging of splenic volume and flow cytometry of CD5+CD19+ cells during the treatment phase revealed that acalabrutinib monotherapy was superior to the combinatorial therapy. Even more surprising was the fact that while anti-PD-1 and acalabrutinib monotherapies provided an improvement in survival compared to vehicle treatment (p = 0.05 and p < 0.0001, respectively), the combination of these two agents actually diminished overall survival (p = 0.77). Shockingly, histopathological analyses of tumors from these acalabrutinib+anti-PD-1 treated mice revealed a significant increase in lymph node involvement and tumors with a high mitotic index. Consistent with this, we observed that treatment of CD5+CD19+ tumor cells from Eµ-TCL1 transgenic mice in vitro with anti-PD-1 antibody plus acalabrutinib markedly increased the proliferative index as measured by EdU incorporation assay compared to acalabrutinib treated group. To understand the molecular events responsible for the observed acalabrutinib+anti-PD-1 hyperproliferative phenotypes, we are examining complementary pathways that may be aberrantly activated/repressed by this combination. Currently, we have identified expression changes in both the NFkb and PKC signaling pathways when these agents are used in combination, and are working to understand how this combination may impact tumor progression. We are also assessing the possibility that anti-PD-1 interferes with the anti-tumorigenic effect of acalabrutinib by enhancing the BCR signaling pathway in CLL tumor cells. In summary, our results demonstrate that the selective BTK inhibitor, acalabrutinib greatly enhances the survival of Eµ-TCL1 mice compared to either vehicle or ibrutinib alone. Surprisingly, we found that the combination of anti-PD-1 antibody + acalabrutinib therapy is actually detrimental in this CLL model and results in a hyperprogressor phenotype. Therefore, results from ongoing clinical trials evaluating combination strategies of anti-PD-1 antibody therapy plus BTK inhibitor need to be analyzed carefully to ensure that this combination is not leading to a similar hyperprogressor phenotype in patients. The results provided here offer some initial insights into the potential mechanisms of the "hyperprogressive" phenotypes following anti-PD-1 treatment and may highlight pathways that could be useful in blocking these deleterious effects. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 ◽  
Author(s):  
Emily J. Lelliott ◽  
Grant A. McArthur ◽  
Jane Oliaro ◽  
Karen E. Sheppard

The recent advent of targeted and immune-based therapies has revolutionized the treatment of melanoma and transformed outcomes for patients with metastatic disease. The majority of patients develop resistance to the current standard-of-care targeted therapy, dual BRAF and MEK inhibition, prompting evaluation of a new combination incorporating a CDK4/6 inhibitor. Based on promising preclinical data, combined BRAF, MEK and CDK4/6 inhibition has recently entered clinical trials for the treatment of BRAFV600 melanoma. Interestingly, while BRAF- and MEK-targeted therapy was initially developed on the basis of potent tumor-intrinsic effects, it was later discovered to have significant immune-potentiating activity. Recent studies have also identified immune-related impacts of CDK4/6 inhibition, though these are less well defined and can be both immune-potentiating and immune-inhibitory. BRAFV600 melanoma patients are also eligible to receive immunotherapy, specifically checkpoint inhibitors against PD-1 and CTLA-4. The immunomodulatory activity of BRAF/MEK-targeted therapies has prompted interest in combination therapies incorporating these with immune checkpoint inhibitors, however recent clinical trials investigating this approach have produced variable results. Here, we summarize the immunomodulatory effects of BRAF, MEK and CDK4/6 inhibitors, shedding light on the prospective utility of this combination alone and in conjunction with immune checkpoint blockade. Understanding the mechanisms that underpin the clinical efficacy of these available therapies is a critical step forward in optimizing novel combination and scheduling approaches to combat melanoma and improve patient outcomes.


Sign in / Sign up

Export Citation Format

Share Document