scholarly journals A CpG-loaded tumor cell vaccine induces antitumor CD4+ T cells that are effective in adoptive therapy for large and established tumors

Blood ◽  
2011 ◽  
Vol 117 (1) ◽  
pp. 118-127 ◽  
Author(s):  
Matthew J. Goldstein ◽  
Bindu Varghese ◽  
Joshua D. Brody ◽  
Ranjani Rajapaksa ◽  
Holbrook Kohrt ◽  
...  

Abstract We designed a whole tumor cell vaccine by “loading” lymphoma tumor cells with CG-enriched oligodeoxynucleotide (CpG), a ligand for the Toll-like receptor 9 (TLR9). CpG-loaded tumor cells were phagocytosed, delivering both tumor antigen(s) and the immunostimulatory CpG molecule to antigen-presenting cells (APCs). These APCs then expressed increased levels of costimulatory molecules and induced T-cell immunity. TLR9 was required in the APCs but not in the CpG-loaded tumor cell. We demonstrate that T cells induced by this vaccine are effective in adoptive cellular therapy for lymphoma. T cells from vaccinated mice transferred into irradiated, syngeneic recipients protected against subsequent lymphoma challenge and, remarkably, led to regression of large and established tumors. This therapeutic effect could be transferred by CD4+ but not by CD8+ T cells. A CpG-loaded whole-cell vaccination is practical and has strong potential for translation to the clinical setting. It is currently being tested in a clinical trial of adoptive immunotherapy for mantle-cell lymphoma.

2020 ◽  
Vol 38 (5_suppl) ◽  
pp. 77-77
Author(s):  
Ganapathy Sriram ◽  
Lauren Milling ◽  
Jung-Kuei Chen ◽  
Wuhbet Abraham ◽  
Darrell J. Irvine ◽  
...  

77 Background: Immune checkpoint inhibition or ICI (antibodies to PD-1 and CTLA4), has shown promise in the treatment of some tumor types, especially in inducing durable remissions in advanced stage cancer patients. However, the majority of patients do not respond to ICI. Identifying combinations to enhance response to ICI is an urgent medical need. Methods: Murine tumor cell lines B16-Ova and MC-38-Ova were treated with DNA-damaging chemotherapeutic drugs, co-cultured with primary murine bone marrow derived dendritic cells (BMDC) followed by addition of OT-1 CD8+ T-cells and flow cytometric analysis of IFN-γ+ CD8+ T-cells. Mice bearing B16-Ova or MC-38 flank tumors were injected intra-tumorally with ex vivo chemotherapy-treated B16-Ova cells with or without systemic ICI. Tumor cross-sectional area was measured using calipers. Intra-tumoral DC and circulating H2-Kb/SIINFEKL-specific CD8+ T-cells were analyzed by flow cytometry. Results: Etoposide and mitoxantrone-treated B16-Ova and MC-38-Ova tumor cells, when co-cultured with BMDC, efficiently promote IFN- γ induction in OT-1 CD8+ T-cells. This was abrogated by co-treatment of tumor cells with Necrostatin-1 but not ZVAD-FMK. Intra-tumoral injection of ex vivo etoposide-treated tumor cells, with systemic ICI, increases the numbers of intra-tumoral CD103+ DC, the frequency of circulating H2-Kb/SIINFEKL-specific CD8+ T-cells and significantly improves survival. The tumor cell vaccine/systemic ICI combination, but not ICI alone, induced complete tumor regressions in a subset of mice. This is abrogated in BATF3-deficient mice. Conclusions: Etoposide and mitoxantrone-treated tumor cells efficiently promote BMDC-mediated CD8+ T-cell priming, in a tumor cell RIPK1 activity-dependent but caspase-independent manner. Intra-tumoral administration of the DNA-damage induced tumor cell vaccine in vivo, in combination with systemic ICI, enhances anti-tumor CD8+ T-cell responses, tumor-free and overall survival, and anti-tumor immunological memory. This enhancement in therapeutic efficacy is dependent on BATF3+ DC in vivo.


2004 ◽  
Vol 227 (2) ◽  
pp. 93-102 ◽  
Author(s):  
Sybren L. Meijer ◽  
Annemieke Dols ◽  
Hong-Ming Hu ◽  
Yiwei Chu ◽  
Pedro Romero ◽  
...  

Vaccine ◽  
2017 ◽  
Vol 35 (43) ◽  
pp. 5932-5938 ◽  
Author(s):  
Lijun Mo ◽  
Qianmei Chen ◽  
Xinji Zhang ◽  
Xiaojun Shi ◽  
Lili Wei ◽  
...  

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 929-929
Author(s):  
Matthew J Goldstein ◽  
Bindu Varghese ◽  
Ranjani Rajapaksa ◽  
Joshua Brody ◽  
Shoshana Levy ◽  
...  

Abstract Abstract 929 Background: Recently, we have investigated adoptive cell therapy for treating lymphoma. The efficacy of this maneuver has been demonstrated by curing large established tumors. Specifically, we use active immunization to generate anti-tumor T cells in vivo and transfer these T cells into lymphodepleted recipient mice (Brody J, Goldstein MJ, Czerwinski DK, and Levy R; Blood, 2009). A major challenge in adoptive therapy is the method of generating anti-tumor T cells. Traditionally, tumor-specific T cells are expanded to large numbers ex vivo. Herein, we describe a new, whole-cell vaccine that is effective in inducing anti-tumor T cells in vivo. This vaccine combines tumor antigens with an immune stimulant: irradiated-tumor cells (a source of tumor antigens) are loaded with the TLR agonist CpG (an immune stimulant). Our vaccine approach has several potential advantages: (1) anti-tumor immunity generated by our CpG-loaded, whole-cell vaccine is poly-antigenic and thus, not limited by the expression of a single antigen target on tumor cells; (2) ex vivo expansion may generate large numbers of effector T cells that can induce tumor regression in the short-term, but have a limited ability to maintain a persistent anti-tumor response. Our model avoids ex vivo manipulation of anti-tumor T cells and thus, may preserve and enhance a memory T cell population that sustains the anti-tumor response. Methods: We derived a new pre-B cell lymphoma cell line in the C57BL/6 background. Primary bone marrow cells were isolated from C57BL/6 donor mice and transfected with a recombinant retrovirus containing the Bcr-Abl oncogene. The emerging transformed cell line was designated H11. This cell line expressed the B lineage marker CD19 but was negative for MHC II and surface Ig. Irradiated H11 tumor cells were pre-loaded with CpG for 24 hours and administered to donor mice by daily, sub-cutaneous injections for five days. Donor splenocytes were harvested seven days following vaccination and adoptively transferred into lethally irradiated recipient mice that were subsequently challenged with a lethal dose of H11 tumor cells. Results: Vaccination with CpG-loaded H11 tumor cells (CpG-H11) generated anti-tumor T cells that are effective in adoptive cell therapy. 100% of mice receiving adoptive therapy with vaccine-induced T cells were protected from tumor challenge. In contrast, vaccination of donor mice with untreated H11 tumor was insufficient for generating anti-tumor T cells. Only 20% of mice treated with T cells from these donors were protected from tumor challenge. In spite of the H11 tumor being MHC Class II−, we observed that anti-tumor immunity generated by the CpG-H11 vaccine was CD4 T cell mediated. CD4 T cells were isolated from CpG-H11 vaccinated donors by flow cytometry. Fewer than 1.8×106 CD4 T cells were sufficient to protect 80% of recipient mice from tumor challenge. In contrast, equivalent numbers of donor CD8 T cells provided no benefit. These results strongly suggest that the CpG-H11 vaccine induced cross-presentation of tumor antigens by antigen-presenting cells (APCs). We have demonstrated that CpG-loaded H11 tumor cells can leak CpG into the immediate environment activating nearby APCs. These APCs have greater phagocytic potential and express higher levels of co-stimulatory molecules such as CD40. Ongoing studies will determine whether APCs which encounter the CpG-H11 vaccine but not untreated H11 tumor cells, can stimulate proliferation of anti-tumor T cells. Conclusions: Here we describe a novel, whole-cell vaccine approach that induces anti-tumor T cells for adoptive therapy to treat lymphoma. This vaccine is superior to vaccination with tumor cells alone. We are currently developing this therapy for evaluation in a clinical trial to treat mantle cell lymphoma. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1997 ◽  
Vol 90 (2) ◽  
pp. 549-561 ◽  
Author(s):  
Angelo A. Cardoso ◽  
Mark J. Seamon ◽  
Hernani M. Afonso ◽  
Paolo Ghia ◽  
Vassiliki A. Boussiotis ◽  
...  

Abstract In contrast to other neoplasms, antigen-specific autologous cytolytic T cells have not been detected in patients with human pre-B–cell leukemias. The absence of efficient B7 family (B7-1/CD80; B7-2/CD86) -mediated costimulation has been shown to be a major defect in tumor cells' capacity to function as antigen-presenting cells. We show here the generation of autologous anti–pre-B–cell leukemia-specific cytolytic T-cell lines from the marrows of 10 of 15 patients with pre-B–cell malignancies. T-cell costimulation via CD28 is an absolute requirement for the generation of these autologous cytolytic T cells (CTL). Although costimulation could be delivered by either bystander B7 transfectants or professional antigen-presenting cells (indirect costimulation), optimal priming and CTL expansion required that the costimulatory signal was expressed by the tumor cell (direct costimulation). These anti–pre-B–cell leukemia-specific CTL lysed both unstimulated and CD40-stimulated tumor cells from each patient studied but did not lyse either K562 or CD40-stimulated allogeneic B cells. Cytolysis was mediated by the induction of tumor cell apoptosis by CD8+ T cells via the perforin-granzyme pathway. Although we were able to generate anti–leukemia-specific CTL from the bone marrow, we were unable to generate such CTL from the peripheral blood of these patients. These studies show that antigen-specific CTL can be generated from the bone marrow of patients with pre-B–cell leukemias and these findings should facilitate the design of adoptive T-cell–mediated immunotherapy trials for the treatment of patients with B-cell precursor malignancies.


Blood ◽  
2003 ◽  
Vol 101 (5) ◽  
pp. 1718-1726 ◽  
Author(s):  
Raphaël F. Rousseau ◽  
Ann E. Haight ◽  
Charlotte Hirschmann-Jax ◽  
Eric S. Yvon ◽  
Donna R. Rill ◽  
...  

In murine models, transgenic chemokine–cytokine tumor vaccines overcome many of the limitations of single-agent immunotherapy by producing the sequence of T-cell attraction followed by proliferation. The safety and immunologic effects of this approach in humans were tested in 21 patients with relapsed or refractory neuroblastoma. They received up to 8 subcutaneous injections of a vaccine combining lymphotactin (Lptn)– and interleukin-2 (IL-2)–secreting allogeneic neuroblastoma cells in a dose-escalating scheme. Severe adverse reactions were limited to reversible panniculitis in 5 patients and bone pain in 1 patient. Injection-site biopsies revealed increased cellularity caused by infiltration of CD4+ and CD8+ lymphocytes, eosinophils, and Langerhans cells. Systemically, the vaccine produced a 2-fold (P = .035) expansion of CD4+ T cells, a 3.5-fold (P = .039) expansion of natural killer (NK) cells, a 2.1-fold (P = .014) expansion of eosinophils, and a 1.6-fold (P = .049) increase in serum IL-5. When restimulated in vitro by the immunizing cell line, T cells collected after vaccination showed a 2.3-fold increase (P = .02) of T-helper (TH2)–type CD3+IL-4+cells. Supernatant collected from restimulated cells showed increased amounts of IL-4 (11.4-fold; P = .021) and IL-5 (8.7-fold;P = .002). Six patients had significant increases in NK cytolytic activity. Fifteen patients made immunoglobulin G (IgG) antibodies that bound to the immunizing cell line. Measurable tumor responses included complete remission in 2 patients and partial response in 1 patient. Hence, allogeneic tumor cell vaccines combining transgenic Lptn with IL-2 appear to have little toxicity in humans and can induce an antitumor immune response.


Blood ◽  
1997 ◽  
Vol 90 (2) ◽  
pp. 549-561 ◽  
Author(s):  
Angelo A. Cardoso ◽  
Mark J. Seamon ◽  
Hernani M. Afonso ◽  
Paolo Ghia ◽  
Vassiliki A. Boussiotis ◽  
...  

In contrast to other neoplasms, antigen-specific autologous cytolytic T cells have not been detected in patients with human pre-B–cell leukemias. The absence of efficient B7 family (B7-1/CD80; B7-2/CD86) -mediated costimulation has been shown to be a major defect in tumor cells' capacity to function as antigen-presenting cells. We show here the generation of autologous anti–pre-B–cell leukemia-specific cytolytic T-cell lines from the marrows of 10 of 15 patients with pre-B–cell malignancies. T-cell costimulation via CD28 is an absolute requirement for the generation of these autologous cytolytic T cells (CTL). Although costimulation could be delivered by either bystander B7 transfectants or professional antigen-presenting cells (indirect costimulation), optimal priming and CTL expansion required that the costimulatory signal was expressed by the tumor cell (direct costimulation). These anti–pre-B–cell leukemia-specific CTL lysed both unstimulated and CD40-stimulated tumor cells from each patient studied but did not lyse either K562 or CD40-stimulated allogeneic B cells. Cytolysis was mediated by the induction of tumor cell apoptosis by CD8+ T cells via the perforin-granzyme pathway. Although we were able to generate anti–leukemia-specific CTL from the bone marrow, we were unable to generate such CTL from the peripheral blood of these patients. These studies show that antigen-specific CTL can be generated from the bone marrow of patients with pre-B–cell leukemias and these findings should facilitate the design of adoptive T-cell–mediated immunotherapy trials for the treatment of patients with B-cell precursor malignancies.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e14542-e14542
Author(s):  
Meihua Chen ◽  
Jin Yi Lang ◽  
Tao Li ◽  
Qifeng Wang ◽  
Guangchao Xu ◽  
...  

e14542 Background: Immunosuppressive factors in the tumor microenvironment reduce the therapeutic efficacy of cancer vaccines; therefore, dampening the tumor immunosuppressive environment while activating antitumor immunity should be a useful approach for cancer therapy. Cancer-associated fibroblasts (CAFs) are among the most important cellular components of the tumor microenvironment and play an important role in the development and progression of tumors. Fibroblast activation protein α (FAPα) is a type II transmembrane protein specifically expressed by CAFs in most epithelial cancers. Gene sequence homology between human and murine FAPα is 90%. Heterogeneity of genes in the evolution of the formation of such nuances can be utilized to break immune tolerance, enhance immunogenicity, induce tumour cell autoimmune responses and thus inhibit tumor growth. Methods: Tumor cells were transfected with human FAPα plasmids employing the cationic lipid DOTAP. Its antitumor effects were investigated in three established tumor models. The potential immune mechanisms were investigated through adoptive immunotherapy and 51Cr release assay. The distributions of the immune cells in the tumor microenvironment were detected by immunohistochemical staining and flow cytometry. Results: Our results shown that whole tumor cell vaccine expressing human FAPα significantly inhibit tumor growth and prolong the survival of tumor bearing mice. This antitumor immune response was involved both of cellular and humoral immune responses. FAPα specific neutralizing antibodies were found in the serum of vaccinated mice and CAFs were significantly reduced within the tumors. Furthermore, this vaccine enhanced the infiltration of CD4+ T cells and CD8+ T cells, and suppressed the accumulation of immunosuppressive cells in the tumor microenvironment. Conclusions: These findings suggest that whole tumor cell vaccine expressing human FAPα inhibit tumor growth by producing FAPα specific neutralizing antibodies and CTLs, and targeting tumor cells and CAFs. These observations provide a new strategy for the clinical use of genetically modified tumor cells as cancer vaccines.


Sign in / Sign up

Export Citation Format

Share Document