Cancer Vaccines

1999 ◽  
Vol 17 (3) ◽  
pp. 1047-1047 ◽  
Author(s):  
Tim F. Greten ◽  
Elizabeth M. Jaffee

It has been more than 100 years since the first reported attempts to activate a patient's immune system to eradicate developing cancers. Although a few of the subsequent vaccine studies demonstrated clinically significant treatment effects, active immunotherapy has not yet become an established cancer treatment modality. Two recent advances have allowed the design of more specific cancer vaccine approaches: improved molecular biology techniques and a greater understanding of the mechanisms involved in the activation of T cells. These advances have resulted in improved systemic antitumor immune responses in animal models. Because most tumor antigens recognized by T cells are still not known, the tumor cell itself is the best source of immunizing antigens. For this reason, most vaccine approaches currently being tested in the clinics use whole cancer cells that have been genetically modified to express genes that are now known to be critical mediators of immune system activation. In the future, the molecular definition of tumor-specific antigens that are recognized by activated T cells will allow the development of targeted antigen-specific vaccines for the treatment of patients with cancer.

2019 ◽  
Vol 8 (2) ◽  
pp. 227-237 ◽  
Author(s):  
Alexandra E. Turley ◽  
Joseph W. Zagorski ◽  
Rebekah C. Kennedy ◽  
Robert A. Freeborn ◽  
Jenna K. Bursley ◽  
...  

The purpose of this study was to determine the effect of subchronic, oral, low-dose cadmium exposure (32 ppm over 10 weeks) on the rat immune system. We found that cadmium exposure increased the induction of IFNγ and IL-10 in T cells activated ex vivo after cadmium exposure.


2004 ◽  
Vol 8 (2_suppl) ◽  
pp. 1-2
Author(s):  
Daniel N. Sauder

Psoriasis is an immune-mediated skin disease in which T cells initiate and maintain the pathogenic process.1 T cells become activated, migrate into the skin, and induce the keratinocyte proliferation associated with the psoriatic phenotype. The activated T cells that infiltrate the skin express the memory phenotype (CD45RO+).2,3 Both CD4+ and CD8+ memory T-cell subtypes are believed to play a role in the pathogenesis of psoriasis. The effectiveness of many traditional therapies for psoriasis (e.g., cyclosporine, methotrexate, psoralen/ultraviolet A light) can be attributed, at least in part, to the potent immunosuppressive effects of these treatments.4,5 Unfortunately, a lack of selective targeting of the immune system by these therapies may result in treatment-limiting side effects.


2021 ◽  
Author(s):  
Anna H.E. Roukens ◽  
Marion König ◽  
Tim Dalebout ◽  
Tamar Tak ◽  
Shohreh Azimi ◽  
...  

AbstractThe immune system plays a major role in Coronavirus Disease 2019 (COVID-19) pathogenesis, viral clearance and protection against re-infection. Immune cell dynamics during COVID-19 have been extensively documented in peripheral blood, but remain elusive in the respiratory tract. We performed minimally-invasive nasal curettage and mass cytometry to characterize nasal immune cells of COVID-19 patients during and 5-6 weeks after hospitalization. Contrary to observations in blood, no general T cell depletion at the nasal mucosa could be detected. Instead, we observed increased numbers of nasal granulocytes, monocytes, CD11c+ NK cells and exhausted CD4+ T effector memory cells during acute COVID-19 compared to age-matched healthy controls. These pro-inflammatory responses were found associated with viral load, while neutrophils also negatively correlated with oxygen saturation levels. Cell numbers mostly normalized following convalescence, except for persisting CD127+ granulocytes and activated T cells, including CD38+ CD8+ tissue-resident memory T cells. Moreover, we identified SARS-CoV-2 specific CD8+ T cells in the nasal mucosa in convalescent patients. Thus, COVID-19 has both transient and long-term effects on the immune system in the upper airway.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4021-4021
Author(s):  
Paraskevi Diamanti ◽  
Charlotte V. Cox ◽  
Benjamin C Ede ◽  
Penka S. Petrova ◽  
Robert A. Uger ◽  
...  

Abstract Identifying suitable therapeutic targets in childhood B cell precursor acute lymphoblastic leukaemia (BCP ALL) to develop more specific, less toxic, therapies is an ongoing challenge. CD200 (OX-2) is a type-1 membrane glycoprotein that is overexpressed in many haematological malignancies, including ALL. We have previously shown that CD200 antigen levels were significantly higher (~60%) in BCP ALL compared to normal bone marrow cells (0.1%). In addition, in low risk minimal residual disease (MRD) samples, only CD200+ cells could initiate leukaemia in NOD.Cg-Prkdcscid Il2rgtm1Wjl/Sz (NSG) mice, suggesting that CD200 may have a role in ALL initiation and progression in these cases. CD200 is also a key immunosuppressive molecule. When bound to its receptor (CD200R), found on monocytes/macrophages and some T cells, it suppresses the immune system by inhibiting the action of the CD200R-bearing cells. Anti-CD200 antibodies (Ab) exert their effect by binding to CD200 antigen and blocking its interaction with CD200R and not by directly inducing cell death. Treating chronic lymphocytic leukaemia cells with anti-CD200 Abs has been shown to prevent engraftment in NSG mice, demonstrating their potential for therapy in haematological cancers that overexpress CD200. The aim of this study was to investigate the effects of monoclonal anti-CD200 Abs on the viability and functional capacity of childhood BCP ALL cells. Primary BCP ALL samples, expressing varying levels of CD200, were randomly selected and the effects of anti-CD200 Abs were assessed in a mixed lymphocyte reaction (MLR). In parallel, the production of interleukin (IL)-2 was measured using ELISA, as an indicator of immune system activation. Monocytes were isolated from blood of healthy donors and cultured for 7 days with 50ng/ml macrophage-colony stimulating factor. At day 7, macrophages were mixed 1:1 with BCP ALL cells with or without anti-CD200 Ab. CD4+ T cells from the same normal donor were added to the plates after 2 hours in a 1:5 ratio (macrophages : T cells). MLR plates were incubated for 72 hours at 37°C, then supernatants analysed by ELISA for IL-2 production and cells were stained with propidium iodide for flow cytometric analyses. When anti-CD200 was added to BCP ALL cells, the amount of IL-2 produced increased significantly. In cases with high CD200 expression (87 - 93.4%) a 16-18.4 fold increase in IL-2 was observed. In cases with very low expression (<0.9% CD200+) only a 3.2 fold increase was observed, as might be expected. Antibody treatment reduced cell viability by 5-7% in cases with high CD200 expression and by 5% in those with low levels. As a more relevant measure of toxicity, the effects of anti-CD200 Ab were investigated in vivo using 2 MRD low and 2 MRD risk cases. Unsorted cells and both CD200+ and CD200- subpopulations were inoculated into NSG mice and once human leukaemia levels in PB were ≥0.1%, animals received 4 doses of anti-CD200 (20mg/kg i.v.) over 10 days. In low risk cases, a 13.5±24.6 fold reduction in leukaemia burden was observed after only 2 doses, while leukaemia levels in the placebo treated group increased by 5.2±9 fold. Similar effects were observed in mice engrafted with CD200+ cells, with a 1.5±16 fold reduction in leukaemia burden in the Ab treated mice while leukaemia levels increased 4.8±3.8 fold in the placebo group. NSG mice engrafted with MRD risk cases were also treated to assess whether this approach would only apply to low risk cases, which our previous functional analyses had indicated. Engraftment with all inoculated populations was rapid and Ab treatment did not delay progression or reduce disease burden, confirming other approaches will be required for MRD risk cases. Since many therapeutics have adverse side effects, including cardiac toxicity, we investigated the effects of anti-CD200 Abs on primary cardiac myocytes in vitro. The antibodies had no effect on viability or IL-2 production compared to controls. In conclusion, these are very promising results for use of anti-CD200 Abs to treat low risk BCP ALL and further in vivo investigation in a larger cohort of patients is warranted. Disclosures Petrova: Trillium Therapeutics Inc: Employment, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties. Uger:Trillium Therapeutics: Employment, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties.


2019 ◽  
Author(s):  
Miguel A Galván Morales ◽  
Raúl Barrera Rodríguez ◽  
Julio R. Santiago Cruz ◽  
Luis M Terán Juárez

Author(s):  
Erin B Taylor ◽  
Eric M George ◽  
Michael J. Ryan ◽  
Michael R Garrett ◽  
Jennifer M. Sasser

The pregnant Dahl salt-sensitive (S) rat is an established pre-clinical model of superimposed spontaneous preeclampsia characterized by exacerbated hypertension, increased urinary protein excretion, and increased fetal demise. Because of the underlying immune system dysfunction present in preeclamptic pregnancies in humans, we hypothesized that the pregnant Dahl S rat would also have an altered immune status. Immune system activation was assessed during late pregnancy in the Dahl S model and compared to healthy pregnant Sprague Dawley (SD) rats subjected to either a sham procedure or a procedure to reduce uterine perfusion pressure (RUPP). Circulating immunoglobulin and cytokine levels were measured by ELISA and Milliplex bead assay, respectively, and percentages of circulating, splenic, and placental immune cells were determined using flow cytometry. The pregnant Dahl S rat exhibited an increase in CD4+ T cells, and specifically TNFα+CD4+ T cells, in the spleen compared to virgin Dahl S rats. The Dahl also had increased neutrophils and decreased B cells in the peripheral blood as compared to Dahl-virgin rats. SD rats that received the RUPP procedure had increases in circulating monocytes and increased IFN-ɣ+CD4+ splenic T cells. Together these findings suggest that dysregulated T cell activity are important factors in both the pregnant Dahl S rats and SD rats after the RUPP procedure.


2020 ◽  
Vol 16 (1) ◽  
pp. 62-69 ◽  
Author(s):  
Kawalpreet Kaur ◽  
Gopal L. Khatik

Background:: Cancer immunotherapy is a type of cancer treatment which effectively harnesses the natural ability of the immune system to fight against cancer cells. This approach takes into consideration the fact that cancer cells express various types of antigens on their surface. Such tumor antigens can be detected by the immune system. However, cancer cells normally develop resistance to the defensive mechanisms presented by the immune system. Thus, cancer immunotherapy has some challenges in its path but due to its impressive clinical effectiveness, it is considered as the potential and effective mode of treatment for cancer. Methods:: We searched the scientific database using cancer, immunotherapy, and tumor antigens as the keywords. Herein, only peer-reviewed research articles were collected which were useful to our current work. Results:: Cells responsible for incurring natural immunity to the body are engineered in such a way that they become able to efficiently recognize and bind to tumor antigens. Such type of immunotherapy is referred to as active immunotherapy. Another type is passive immunotherapy, which involves the process of modifying the existing natural immune responses against cancer cells. A hybrid type of immunotherapy has also been developed which involves the combinative use of both active and passive immunotherapy. Cancer immunotherapy has so far proven to be an effective treatment for cancer as this therapy primarily aims at attacking cancer cells and not the healthy body cells lying in close vicinity to them. Conclusion:: In the review, we described the significance of immunotherapy in the management of various types of cancer.


Cancers ◽  
2021 ◽  
Vol 14 (1) ◽  
pp. 155
Author(s):  
Soha Bazyar ◽  
Edward Timothy O’Brien ◽  
Thad Benefield ◽  
Victoria R. Roberts ◽  
Rashmi J. Kumar ◽  
...  

Spatially fractionated radiotherapy has been shown to have effects on the immune system that differ from conventional radiotherapy (CRT). We compared several aspects of the immune response to CRT relative to a model of spatially fractionated radiotherapy (RT), termed microplanar radiotherapy (MRT). MRT delivers hundreds of grays of radiation in submillimeter beams (peak), separated by non-radiated volumes (valley). We have developed a preclinical method to apply MRT by a commercial small animal irradiator. Using a B16-F10 murine melanoma model, we first evaluated the in vitro and in vivo effect of MRT, which demonstrated significant treatment superiority relative to CRT. Interestingly, we observed insignificant treatment responses when MRT was applied to Rag−/− and CD8-depleted mice. An immuno-histological analysis showed that MRT recruited cytotoxic lymphocytes (CD8), while suppressing the number of regulatory T cells (Tregs). Using RT-qPCR, we observed that, compared to CRT, MRT, up to the dose that we applied, significantly increased and did not saturate CXCL9 expression, a cytokine that plays a crucial role in the attraction of activated T cells. Finally, MRT combined with anti-CTLA-4 ablated the tumor in half of the cases, and induced prolonged systemic antitumor immunity.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2390-2390
Author(s):  
Kristen Snyder ◽  
Hua Zhang ◽  
Barbara Vance ◽  
Carl H. June ◽  
Robert Vonderheide ◽  
...  

Abstract Background: Most tumor antigens are self antigens, therefore an effective immune response to cancer must break self tolerance. Current models hold that lymphopenia results in proliferation toward self antigens. We sought to determine whether the frequency of T cells specific for self antigens, both tumor associated (TA) and non-tumor associated (NTA) were increased in cancer patients, before or after the induction of lymphopenia. Further, we sought to expand T cells specific for self antigens using artificial APCs which signal via CD3 and 4-1BB. Methods: Using a broad panel of HLA-A2 tetramers, we used flow cytometry to enumerate circulating CD8+ T cells which recognize viral (CMV, EBV, flu), NTA self antigens (PR-1, WT1, MART-1 and CEA) and TA self antigens (NY-ESO1, PRAME, Ofa/iLRP, CYP239/190, Survivin and hTERT) in normal donors (ND) (n=13), cancer patients with Ewings sarcoma studied prior to chemotherapy (n=7) and following cytotoxic chemotherapy (n=5). To exclude non-specific tetramer binding, two tetramers with differing fluorochromes were included in each tube and cells simultaneously binding both tetramers were excluded as non-specific and binding frequency to control tetramers was subtracted. Results: The frequency of T cells with specificity for viral antigens (range 0.0%–0.7%) were similar between ND and cancer patients, with no significant increase seen following lymphopenia. For NTA self antigens frequencies prior to chemotherapy were similar between ND and cancer patients respectively, with the lowest mean frequency seen in CEA tetramer specific cells (0.084% ND vs. 0.088% pts), then PR-1 (0.19% ND vs. 0.019% pts), then WT-1 (0.43% ND vs. 0.20% pts) then MART-1 CD8+ specific tetramer cells (0.82% ND vs. 0.83% pts). Remarkably, the frequency of MART-1 specific cells approximates that seen for CMV in both patients and normal donors. For TA self antigens, patients showed trends toward increased frequencies compared to ND but these were not statistically significant. Following lymphopenia, there was no consistent or statistically significant increase in the frequency of cells with specificity for either TA or NTA self antigens. In an attempt to increase the frequency of tumor reactive T cells for potential use in adoptive cellular immunotherapy, artificial APCs were used to deliver a signal via CD3 and 4-1BB and changes in antigen specificity via tetramers were monitored. This method consistently increased the frequency of viral and self antigen specific (TA and NTA) cells with some cultures demonstrating frequencies for individual TA self antigens which approached 10%. Conclusions: 1. Both normal donors and cancer patients demonstrate sizable repertoires of self-reactive T cells, with the frequency of MART-1 specific cells similar to that seen toward CMV. 2. Patients with cancer retain cells which recognize tumor antigens. However, cells recognizing TA self antigens are only marginally increased compared to that found in normal individuals, and are found at frequencies similar to NTA self antigens implicating inefficient priming by the tumor. 3. Lymphopenia does not significantly augment the frequency of cells responding to TA or NTA self antigens. 4. Sizable expansions of T cells responding to TA self antigens can be induced using CD3/4-1BB based expansion.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3715-3715
Author(s):  
Jacalyn Rosenblatt ◽  
Zekui Wu ◽  
Corrine Lenahan ◽  
Baldev Vasir ◽  
Adam Bissonnette ◽  
...  

Abstract Dendritic Cells (DCs) are potent antigen presenting cells that prominently express costimulatory molecules and are uniquely capable of stimulating primary immune responses. We have developed a promising tumor vaccine involving the fusion of patient derived tumor cells and autologous DCs. However, vaccine efficacy is limited by effector cell dysfunction and increased presence of regulatory T cells characteristic of cancer patients. Ligation of CD3/CD28 has been shown to deliver a powerful antigen-independent stimulus to resting T cell populations. We postulated that the combined exposure to antiCD3/CD28 and DC/tumor fusions would result in the expansion of activated T cells targeting tumor antigens. We have examined the phenotypic and functional characteristics of T cells that have undergone in vitro stimulation with DC/renal carcinoma (RCC) fusion cells in conjunction with expansion using antiCD3/CD28. DCs were generated from adherent mononuclear cells cultured with rhIL-4 and GM-CSF for five days, and matured by 48 hour exposure to TNFa. DCs were fused with RCC by coculture in 50% solution of polyethylene glycol. CD3/CD28 mediated activation was accomplished by culturing cells on plates coated with antiCD3/CD28 antibody for 48 hours. Exposure to fusion cells, antiCD3/CD28 alone, or antiCD3/CD28 followed by DC/tumor fusions resulted in no significant evidence of T cell expansion with a stimulation index (SI) of 0.9, 1.0, and 1.0, respectively. In contrast, a marked synergistic effect on proliferation was observed when T cells underwent stimulation with DC/tumor fusion cells followed by expansion using antiCD3/CD28 (SI 13.2) (p= 0.02, p=0.01, and p= 0.03 compared to anti-CD3/CD28 alone, fusions alone, and antiCD3/CD28 followed by fusion cells, respectively). We assessed the phenotypic characteristics of T cells stimulated by antiCD3/CD28, fusion cells, or their combination. In 10 experiments, stimulation with DC/RCC fusions followed by exposure to antiCD3/CD28 resulted in a nearly 8 fold expansion of CD4+/CD25+ cells (p=0.001 compared to unstimulated T cells). A 16 fold increase in CD4/CD25/CD69+ cells was observed consistent with the dramatic expansion of activated T cells. In contrast, exposure to antiCD3/CD28 alone or antiCD3/CD28 followed by stimulation with fusion cells resulted in a 3 fold expansion of CD4/CD25+ T cells and a modest expansion of CD4/CD25/CD69+ cells. In concert with these findings, IFNγ production by CD4+ T cells was most pronounced (7-fold expansion) following stimulation with DC/tumor fusion vaccine and expansion with anti-CD3/CD28 (p<0.01). We also examined the effect of stimulation with DC/RCC fusions followed by antiCD3/CD28 on the expansion of regulatory T cells. In 9 experiments, stimulation with DC/RCC fusions followed by expansion with antiCD3/CD28 also resulted in a 5-fold and 4.6 fold expansion of CD4/CD25/Foxp3+ and IL-10 expressing T cells, respectively. Stimulation with fusions and antiCD3/CD28 resulted in an increase in CD45RO+ memory effector T cells (2-fold increase, p= 0.08) and a decrease in CD45RA+, naïve T cells. In conclusion, we have shown that stimulation of T cells by DC/RCC fusions followed by exposure to CD3/CD28 antibodies results in the expansion of tumor reactive T cells that predominantly express markers of activation. We are developing a clinical trial in which patients will receive fusion/CD3/CD28 expanded T cells following in vivo depletion of regulatory T cells.


Sign in / Sign up

Export Citation Format

Share Document