scholarly journals Loss of tumor-specific and idiotype-specific immunity with age.

1981 ◽  
Vol 154 (2) ◽  
pp. 275-290 ◽  
Author(s):  
P M Flood ◽  
J L Urban ◽  
M L Kripke ◽  
H Schreiber

The ultraviolet light-induced fibrosarcoma 1591 undergoes "first-set rejection" when transplanted into normal syngeneic mice. We found, however, that the primary resistance of normal mice decreases with age, beginning at 9--12 mo, equivalent to middle age for mice. Mice lose with age the capacity to mount both idiotypic and anti-idiotypic responses responsible for controlling the growth of tumor. This loss was correlated with quantitative as well as qualitative changes in the response, such as changes in specificity and clonotype. Normal young mice regularly expressed a dominant common anti-1591 "idiotype" as defined by an anti-idiotypic probe. The capability of normal mice to respond with lymphocytes of this dominant common idiotype began to decline at about 8 mo of age. At this time, animals still generated tumor-specific lymphocytes, but these lymphocytes appear to be idiotypically different lymphocyte clones. With further increase in age, animals responded with tumor-reactive lymphocytes that showed a marked cross-reactivity to other tumor target cell lines. Both in vivo and in vitro, the capability of normal mice to mount an immune response that was specific for the 1591 tumor cells decreased between 9 and 14 mo, which was the age individual mice became increasingly susceptible to a challenge with 1591 tumor cells. Thus, our data suggest that clones of tumor-specific T cells provide primary and early protection of young animals against challenge with malignant 1591 cells. However, the dominance of these tumor-specific T cell clones in a primary immune response is lost in middle-age. Because the ability of animals to mount anti-idiotypic immune response also declined in middle-aged animals, it is possible that the observed loss of clonal dominance of tumor-specific clones with increasing age is at least partially related to age-dependent changes in the anti-idiotypic compartment.

2019 ◽  
Vol 18 ◽  
pp. 153473541984804 ◽  
Author(s):  
Paola Lasso ◽  
Mónica Llano Murcia ◽  
Tito Alejandro Sandoval ◽  
Claudia Urueña ◽  
Alfonso Barreto ◽  
...  

Background: The tumor cells responsible for metastasis are highly resistant to chemotherapy and have characteristics of stem cells, with a high capacity for self-regeneration and the use of detoxifying mechanisms that participate in drug resistance. In vivo models of highly resistant cells allow us to evaluate the real impact of the immune response in the control of cancer. Materials and Methods: A tumor population derived from the 4T1 breast cancer cell line that was stable in vitro and highly aggressive in vivo was obtained, characterized, and determined to exhibit cancer stem cell (CSC) phenotypes (CD44+, CD24+, ALDH+, Oct4+, Nanog+, Sox2+, and high self-renewal capacity). Orthotopic transplantation of these cells allowed us to evaluate their in vivo susceptibility to chemo and immune responses induced after vaccination. Results: The immune response induced after vaccination with tumor cells treated with doxorubicin decreased the formation of tumors and macrometastasis in this model, which allowed us to confirm the immune response relevance in the control of highly chemotherapy-resistant ALDH+ CSCs in an aggressive tumor model in immunocompetent animals. Conclusions: The antitumor immune response was the main element capable of controlling tumor progression as well as metastasis in a highly chemotherapy-resistant aggressive breast cancer model.


Author(s):  
С.В. Калиш ◽  
С.В. Лямина ◽  
А.А. Раецкая ◽  
О.П. Буданова ◽  
И.Ю. Малышев

Цель - представить доказательства правомерности гипотезы, что комбинированный пул репрограммированных in vitro макрофагов и лимфоцитов будет эффективно ограничивать пролиферацию опухолевых клеток in vitro , а при введении в организм будет существенно ограничивать развитие опухоли in vivo . Методика. Размножение опухолевых клеток инициировали in vitro путем добавления клеток карциномы Эрлиха (КЭ) в среду культивирования RPMI-1640. Развитие асцитной опухоли in vivo воспроизводили путем внутрибрюшной инъекции клеток КЭ мышам. Результаты. Установлено, что M3 макрофаги вместе с антиген-репрограммированными лимфоцитами оказывают выраженный противоопухолевый эффект и in vitro, и in vivo , который был существеннее противоопухолевого эффекта цисплатина. Заключение. Факты, свидетельствующие, что М3 макрофаги в сочетании с in vitro антиген-репрограммированными лимфоцитами значительно подавляют рост опухоли in vivo , делают перспективным разработку клинической версии биотехнологии ограничения роста опухоли путем предварительного программирования противоопухолевого иммунного ответа «в пробирке». Aim. To test a hypothesis that a combined pool of in vitro reprogrammed macrophages and lymphocytes will effectively limit growth of tumor cells in vitro , and injections of these cells into the body will considerably limit development of a tumor in vivo . Methods. Tumor growth was initiated in vitro by addition of Ehrlich carcinoma (EC) cells to the RPMI-1640 cell culture medium and in vivo by intraperitoneal injection of EC cells into mice. Results. M3 macrophages in combination with antigen-reprogrammed lymphocytes exerted a pronounced antitumor effect both in vitro and in vivo, which was superior to the effect of cisplatin. Conclusion. M3 macrophages in combination with in vitro antigen-reprogrammed lymphocytes significantly inhibited the tumor growth in vivo . This fact justifies development of a clinical version of the tumor growth restricting biotechnology using pre-programming of the antitumor immune response in vitro .


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4405-4405
Author(s):  
Eugene Zhukovsky ◽  
Uwe Reusch ◽  
Carmen Burkhardt ◽  
Stefan Knackmuss ◽  
Ivica Fucek ◽  
...  

Abstract To harness the potent tumor-killing capacity of T cells for the treatment of CD19+ malignancies, we developed a humanized bispecific tetravalent antibody, with two binding sites for CD3 and CD19, the CD19/CD3 RECRUIT-TandAb AFM11. CD19 is expressed from early B cell development through differentiation into plasma cells, and is an attractive alternative to CD20 as a target for the development of therapeutic antibodies to treat B cell malignancies such as Non Hodgkin Lymphoma. Since native antibodies cannot recruit T cells, we engineered a bispecific anti-CD19/anti-CD3 TandAb. The tumor-specific CD19 antigen module targets the TandAb to cancer cells, while simultaneously, the CD3 effector module recruits and activates T cells, leading to cancer cell lysis. The advantages of the TandAb technology, relative to other bi-functional fragment antibody scaffolds, include: improved pharmacokinetics (PK) enabling intravenous dosing, more drug-like properties, and avidity-enhanced efficacy for the targeting and killing of tumor cells. We evaluated in vitro efficacy and safety using CD19+ cell lines, and in vivo efficacy in a murine NOD/scid xenograft model reconstituted with human PBMC. Further, we used standard preclinical IND enabling assays to evaluate tissue cross reactivity, PK, and toxicological profile (local tolerance, hematocompatibility, effects on hematopoesis, etc). In vitro assays demonstrated the higher potency and efficacy of target cell lysis by AFM11 relative to a bispecific tandem scFv (that is currently in clinical evaluation). CD8+ T cells dominate early AFM11-mediated cytotoxicity (4 hrs) while after 24 hrs both CD4+ and CD8+ T cells equally contribute to tumor lysis with EC50 between 0.5 – 5 pM; cytotoxicity was independent of CD19 cell-surface density. AFM11 exhibited similar cytotoxicity over effector:target ratios ranging from 5:1 to 1:5, and facilitated serial T cell-killing of its targets. The advantage of AFM11 over the bispecific tandem scFv was most pronounced at lower effector:target ratios. AFM11 activated T cells only in the presence of CD19+ cells. In PBMC cultures, AFM11 induced CD69 and CD25 expression, T cell proliferation, and production of IFN-γ, TNF-α, IL-2, IL-6, and IL-10. Depletion of CD19+ cells from PBMC abrogated these effects, demonstrating that the T cell activation is strictly CD19+ target-dependent. Thus, AFM11 should not elicit the devastating cytokine release observed when full-length antibodies bind CD3. Up to one week co-incubation with AFM11 did not inhibit T cell cytotoxicity, suggesting that the TandAb does not induce anergy. In vivo, AFM11 induced dose-dependent growth inhibition of Raji tumors; a single 0.5 mg/kg dose exhibited efficacy similar to 5 daily injections. In the tissue cross reactivity study, only tissues containing CD19+ and CD3+ cells were stained by AFM11; all other tissues, including vital organs, displayed no cross reactivity. Similarly, no local intolerance was observed in rabbits, and no effect on myeloid and erythroid progenitors was observed in a colony-forming assay. Strong accumulation of 125I-labeled AFM11 was observed in the tumors of mice engrafted with CD19+ cancer cells, and no unspecific organ accumulation was observed. Finally, evaluated on the basis of Cmax and the area under the curve (AUC), AFM11 exhibited dose linearity (20 – 500 mg AFM11 dose range) upon single i.v. bolus administration in mice; half-life (T1/2) ranged from 18.4 to 22.9 hr. In summary, AFM11 is a highly efficacious novel drug candidate for the treatment of CD19+ malignancies with an advantageous safety profile and anticipated dosing regimen. Disclosures: Zhukovsky: Affimed Therapeutics AG: Employment, Equity Ownership. Reusch:Affimed Therapeutics AG: Employment. Burkhardt:Affimed Therapeutics AG: Employment. Knackmuss:Affimed Therapeutics AG: Employment. Fucek:Affimed Therapeutics AG: Employment. Eser:Affimed Therapeutics AG: Employment. McAleese:Affimed Therapeutics AG: Employment. Ellwanger:Affimed Therapeutics AG: Employment. Little:Affimed Therapeutics AG: Consultancy, Equity Ownership.


2020 ◽  
Vol 13 (4) ◽  
pp. 1845-1862
Author(s):  
Leana Rich M. Herrera

The rapid transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted to the death of hundreds of thousands of people worldwide. With the devastating effects on the economy and healthcare system of many countries, it is crucial to acceleratevaccine development against SARS-CoV-2. Thus, thisworkutilizedimmunoinformaticsto efficiently design a novel multi-epitope vaccine that can potentially induce immune response through the immunogenic, and abundantly expressed structural proteins in SARS-CoV-2. Epitopes were screened and evaluated using various immunoinformatics tools and databases. Antigenicity, allergenicity, and population coverage were assessed. Epitopes were adjoined to form a single vaccine construct (Covax),linked with 50S ribosomal protein as an adjuvant. Physicochemical properties, cross-reactivity, antigenicity,andallergenicityof Covax were evaluated. The tertiary structure of Covax was modeled, refined and validated for docking with toll-like receptor 4 (TLR4). Binding affinity of Covax-TLR4 was estimated and compared with TLR4-adjuvant as control. Lastly,the immune response with Covax was simulated and compared withadjuvant alone. Total of 33 epitopes from S (21), E (3), M (5),and N (4)proteins were merged in Covax. These include epitopes on thereceptor-binding motif (RBM) of S protein known to beessential in the viral attachment. In silico evaluations classified Covax as stable, antigenic, and non-allergenic. Epitopes were estimated to have large worldwide population coverage, especially in areas with high infection rates, indicating broad potential efficacy of Covax as a vaccine for the most affected populations.Results in this work showed that Covax can bind to TLR4 whichindicates potential immunogenicity and superior properties necessary for a successful vaccine. Overall, this work efficiently minimized time, effort and cost in designing a candidate vaccine against SARS-CoV-2. In vitro and in vivo studies on Covax are anticipated.


2012 ◽  
Vol 2012 ◽  
pp. 1-9 ◽  
Author(s):  
Xin Yong ◽  
Yü-Feng Xiao ◽  
Gang Luo ◽  
Bin He ◽  
Mu-Han Lü ◽  
...  

Vaccine-induced cytotoxic T lymphocytes (CTLs) play a critical role in adaptive immunity against cancers. An important goal of current vaccine research is to induce durable and long-lasting functional CTLs that can mediate cytotoxic effects on tumor cells. To attain this goal, there are four distinct steps that must be achieved. To initiate a vaccine-induced CTL antitumor immune response, dendritic cells (DCs) must capture antigens derived from exogenous tumor vaccines in vivo or autologous DCs directly loaded in vitro with tumor antigens must be injected. Next, tumor-antigen-loaded DCs must activate CTLs in lymphoid organs. Subsequently, activated CTLs must enter the tumor microenvironment to perform their functions, at which point a variety of negative regulatory signals suppress the immune response. Finally, CTL-mediated cytotoxic effects must overcome the tolerance induced by tumor cells. Each step is a complex process that may be impeded in many ways. However, if these steps happen under appropriate regulation, the vaccine-induced CTL antitumor immune response will be more successful. For this reason, we should gain a better understanding of the basic mechanisms that govern the immune response. This paper, based on the steps necessary to induce an immune response, discusses current strategies for enhancing vaccine-induced CTL antitumor immune responses.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 297-297
Author(s):  
I. Jedema ◽  
E. Steeneveld ◽  
M. Hoogendoorn ◽  
R. Willemze ◽  
J.H.F. Falkenburg

Abstract By CD40 crosslinking in the presence of cytokines leukemic cells can be modified into good antigen presenting cells (APC) expressing costimulatory molecules CD40, CD80, CD86, and CD83. Previously, primary alloreactive T cell responses from HLA-matched donors have been generated using these leukemic-APC as stimulator cells against acute and chronic myeloid leukemia (AML&CML), and acute and chronic lymphocytic leukemias (ALL&CLL). However, the likelihood of generating a good immune response is highly unpredictable and long-term culture in the presence of high dose IL-2 is needed to enrich for leukemia-reactive T cells. Since the length of the in-vitro culture period has been shown to be inversely correlated with the potential of cells to survive and expand in-vivo, we developed a method facilitating early activation, detection and rapid isolation of leukemia-reactive T cells based on their interferon-gamma (IFNg) secretion using the cytokine capture assay (Miltenyi). In order to enrich for leukemia-reactive T cells and to synchronize the production of IFNg, T cells were first stimulated with the leukemic APC with addition of low dose IL-2 (10 IU/mL) at day 7, resulting in re-entry of the majority of the T cells into a quiescent state after 14 days of culture. Then, the cells were specifically restimulated resulting in synchronized production of IFNg and allowing efficient isolation. Using this method we were able to isolate T cell populations containing a high frequency of leukemia-reactive T cells against CLL, ALL, AML, and CML in 11 donor/patient pairs. Using a CFSE-based cytotoxicity assay (Jedema, Blood2004; 103: 2677) as read-out we were able to demonstrate 20–80% lysis of the primary leukemic blasts by the IFNg+ T cells at very low E/T ratios (3/1-0.3/1) in the majority of the responses, whereas the IFNg- fractions induced only 5–30% lysis. Single cell sorting of the IFNg producing T cells revealed that 15–30% of the T cell clones was capable of exerting minor antigen specific cytotoxic activity against the patient cells in an HLA-restricted fashion. However, in individual cases despite minor antigen disparities between donor and patient no specific anti-leukemia immune response could be detected. Prior to exposure to the leukemic-APC in-vivo activated T cells were observed in the responder T cell population of these donors that contained a high frequency of regulatory T cells defined as CD4+/CD25+, CD4+/CD152+, and CD8+/CD28−. We hypothesized that these regulatory T cells might actively inhibit the induction of an anti-leukemic T cell response. Therefore, in a donor/CLL patient pair, in which we were not able to induce a cytotoxic immune response against the CLL-APC we removed the in-vivo activated T cells from the responder material prior to the initial activation with the CLL-APC. Whereas no cytotoxic activity could be isolated from unmodified responder material (only 1/288 clones was cytotoxic), the IFNg+ T cells isolated from the response induced after depletion of the in-vivo activated T cells was capable of exerting massive cytotoxicity against both the primary CLL (55%) and the CLL-APC (70%). Single cell cloning of this response revealed that 35/129 T cell clones (>25%, 26 CD8+, 9 CD4+) exerted HLA-restricted CLL-specific cytotoxicity. From these results we conclude that the likelihood of generating a primary anti-leukemic immune response is not only determined by the frequency of precursor CTLs, but also by the frequency of inhibitory regulatory T cells at the onset of the immune response.


2005 ◽  
Vol 202 (12) ◽  
pp. 1691-1701 ◽  
Author(s):  
Noelia Casares ◽  
Marie O. Pequignot ◽  
Antoine Tesniere ◽  
François Ghiringhelli ◽  
Stéphan Roux ◽  
...  

Systemic anticancer chemotherapy is immunosuppressive and mostly induces nonimmunogenic tumor cell death. Here, we show that even in the absence of any adjuvant, tumor cells dying in response to anthracyclins can elicit an effective antitumor immune response that suppresses the growth of inoculated tumors or leads to the regression of established neoplasia. Although both antracyclins and mitomycin C induced apoptosis with caspase activation, only anthracyclin-induced immunogenic cell death was immunogenic. Caspase inhibition by Z-VAD-fmk or transfection with the baculovirus inhibitor p35 did not inhibit doxorubicin (DX)-induced cell death, yet suppressed the immunogenicity of dying tumor cells in several rodent models of neoplasia. Depletion of dendritic cells (DCs) or CD8+T cells abolished the immune response against DX-treated apoptotic tumor cells in vivo. Caspase inhibition suppressed the capacity of DX-killed cells to be phagocytosed by DCs, yet had no effect on their capacity to elicit DC maturation. Freshly excised tumors became immunogenic upon DX treatment in vitro, and intratumoral inoculation of DX could trigger the regression of established tumors in immunocompetent mice. These results delineate a procedure for the generation of cancer vaccines and the stimulation of anti-neoplastic immune responses in vivo.


Sign in / Sign up

Export Citation Format

Share Document