scholarly journals Adjuvant Antitumor Immunity Contributes to the Overall Antitumor Effect of Pegylated Liposomal Doxorubicin (Doxil®) in C26 Tumor-Bearing Immunocompetent Mice

Pharmaceutics ◽  
2020 ◽  
Vol 12 (10) ◽  
pp. 990
Author(s):  
Takuma Takayama ◽  
Taro Shimizu ◽  
Amr S. Abu Lila ◽  
Yuki Kanazawa ◽  
Hidenori Ando ◽  
...  

Doxorubicin (DXR) has been reported to have direct cytotoxicity against cancer cells and indirect immunotoxicity by modulation of host antitumor immunity. Hence, it may prevent cancer progression by a dual mechanism. Doxil®, a formulation of DXR encapsulated in polyethylene glycol modified (PEGylated) liposomes, is the most widely used of the clinically approved liposomal anticancer drugs. However, the effect of Doxil® on host antitumor immunity is not well understood. In this study, Doxil® efficiently suppressed tumor growth in immunocompetent mice bearing C26 murine colorectal carcinomas, but not in T cell-deficient nude mice, indicating a contribution of T cells to the overall antitumor effect of Doxil®. In immunocompetent mice, Doxil® increased major histocompatibility complex (MHC-1) levels in C26 tumors, which may be an indicator of increased immunogenicity of tumor cells, and potentially amplified tumor immunogenicity by decreasing immunosuppressive cells such as regulatory T cells, tumor-associated microphages and myeloid-derived suppressor cells that collectively suppress T cell-mediated antitumor responses. This suggests that encapsulation of DXR into PEGylated liposomes increased the therapeutic efficacy of DXR though effects on host antitumor immunogenicity in addition to direct cytotoxic effects on tumor cells. This report describes the role of host antitumor immunity in the overall therapeutic effects of Doxil®. Manipulating pharmacokinetics and biodistribution of chemotherapeutic agents with immunomodulatory properties may increase their therapeutic efficacies by amplifying host antitumor immunity in addition to direct cytotoxic effects on tumor cells.

2021 ◽  
Vol 14 (11) ◽  
pp. 1172
Author(s):  
Daisuke Kamakura ◽  
Ryutaro Asano ◽  
Masahiro Yasunaga

As a breakthrough immunotherapy, T cell bispecific antibodies (T-BsAbs) are a promising antibody therapy for various kinds of cancer. In general, T-BsAbs have dual-binding specificity to a tumor-associated antigen and a CD3 subunit forming a complex with the TCR. This enables T-BsAbs to crosslink tumor cells and T cells, inducing T cell activation and subsequent tumor cell death. Unlike immune checkpoint inhibitors, which release the brake of the immune system, T-BsAbs serve as an accelerator of T cells by stimulating their immune response via CD3 engagement. Therefore, they can actively redirect host immunity toward tumors, including T cell recruitment from the periphery to the tumor site and immunological synapse formation between tumor cells and T cells. Although the low immunogenicity of solid tumors increases the challenge of cancer immunotherapy, T-BsAbs capable of immune redirection can greatly benefit patients with such tumors. To investigate the detailed relationship between T-BsAbs delivery and their T cell redirection activity, it is necessary to determine how T-BsAbs deliver antitumor immunity to the tumor site and bring about tumor cell death. This review article discusses T-BsAb properties, specifically their pharmacokinetics, redirection of anticancer immunity, and local mechanism of action within tumor tissues, and discuss further challenges to expediting T-BsAb development.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3246-3246
Author(s):  
Yosuke Yokota ◽  
Hiroyuki Inoue ◽  
Ayumi Watanabe ◽  
Chika Sakamoto ◽  
Megumi Narusawa ◽  
...  

Abstract Abstract 3246 GM-CSF (Granulocyte macrophage-colony stimulating factor) gene-transduced leukemia cell vaccine has been reported to be promising approach to enhance antitumor immune response in leukemia patients. The enhancement of this specific antitumor immunity is considered to be clinically beneficial to maintain complete remission for long time after chemotherapy without stem cell transplantion. Leukotriene B4 (LTB4) is an extremely potent lipid inflammatory mediator derived from membrane phospholipids, known to recruit and activate leukocytes including neutrophils, mediated by the same class of receptors, the GPCR (G-protein coupled receptor) superfamily, BLT1 and BLT2. So far, the role of LTB4 in tumor immunology is not well known. Previously, we demonstrated that the GM-CSF gene transduction into murine monocytic leukemia cell line of WEHI3B (WGM) eliminated the tumorigenicity in subcutaneous challenge model using wild type (WT) BALB/c mice. At day 50 after the challenge, we rechallenged WEHI3B cells into the opposite flank of both WT and BLT1-KO mice which had rejected WGM cells (WT/WGM, KO/WGM, respectively). Intriguingly, more KO/WGM mice re-rejected the rechallenged WEHI3B cells and showed significantly prolonged survival compared with WT/WGM mice. To clarify this unique mechanism of the long-lasting antitumor effects observed in KO/WGM mice, we performed following immunological assays. Our in vivo immune cell depletion assays (NK, CD4+ T, CD8+ T cell) showed that KO/WGM mice treated with anti-CD4 antibody displayed rather enhanced tumor growth compared with WT/WGM mice. Thus, we next compared the rates of different subsets of memory T cells, so called memory stem T cell subsets (TSCM), central memory T cell subsets (TCM) and effector memory T cell subsets (TEM) in TDLNs (Tumor draining lymph nodes) between WT/WGM mice and KO/WGM mice at the day 46 after tumor challenge. Expectedly, results showed that TDLNs harvested from KO/WGM mice exhibited higher subset ratios of CD44+CD62L+ (TCM) cell to CD4+ T cells as well as CD44+CD62L− or (TEM) cell to CD4+ T cells than those from WT/WGM mice. In the case of use of CD122 for memory marker, similar results were obtained. Of note, TDLNs from KO/WGM mice exhibited increased ratios of CD44+CD122+CD62L− (TSCM) cell. In addition, the cell numbers of immunosuppressive CD3+ CD4+ PD-1+, CD3+ CD4+ GITR+ and CD3+ CD4+ CTLA4+ cells were reduced in both TDLNs and spleen derived from KO/WGM mice compared with those from WT/WGM mice. Furthermore, our results of CBA (Cytometric Bead Array) assay using splenocytes harvested from day 2 to day 15 showed that the Th2 cytokine production level of IL-4 and IL-5 from splenocyte harvested from KO/WGM mice were higher than those from WT/WGM mice. In regard to IL-2 and IFN-g (Th1 cytokine), the production level at both day 7 and day10 from splenocytes harvested from KO/WGM mice were also higher than those from WT/WGM mice, implicating that loss of LTB4/BLT1 signaling promotes systemic activation of both tumor antigens specific Th1 and Th2 CD4+ T cell subsets. Finally, our flow cytometric analyses demonstrated that exogenously GM-CSF-driven upregulated MFI of CD40+, CD80+ and CD86+ DCs in TDLNs from KO/WGM mice were further significantly increased compared with those from WT/WGM mice (p<0.05), and more numerous number of CD86+ DCs that had phagocytosed TAAs of WEHI3B cells was detected in TDLNs harvested from KO/WGM mice than that from WT/WGM mice. In conclusion, we for the first time demonstrate that loss of LTB4/BLT1 axis can sustain long-lasting memory CD4+ T cell-dependent antitumor immunity against syngeneic tumor cells long after GM-CSF triggering tumor rejection, allowing us to expect the possibility that the strategy of blocking LTB4/BLT1signaling may be useful to maintain antitumor immunity induced by GM-CSF gene-transduced leukemia cell vaccine in clinical settings. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3172-3172
Author(s):  
Aaron Foster ◽  
Fatma Okur ◽  
Gianpietro Dotti ◽  
Jessica Shafer ◽  
An Lu ◽  
...  

Abstract Side-population (SP) cells, defined on the basis of their ability to rapidly expel Hoechst dye, may be precursor cells for normal and malignant tissues and are more resistant to chemotherapy. We have now characterized a malignant SP subset in the peripheral blood of B-CLL patients and show how vaccination with CD40L/IL-2 expressing autologous tumor cells can induce a T cell immune response that reduces or eliminates this tumor subpopulation. Hoechst 33342 staining combined with CD5/CD19 antibody labeling of peripheral blood mononuclear cells showed a distinct CD5+CD19+ SP phenotype (median=0.22%, range; 0.2–2.17%) in 85% (18/21) of B-CLL patients (Stage I–III). We sorted SP and non-SP (NSP) cells and extracted RNA and performed a comprehensive gene expression analysis by microarray (n=9 donor SP and NSP pairs), finding 85 genes overexpressed (&gt;2-fold log ratio) in SP compared to NSP, and 228 genes higher in NSP versus SP. The analysis showed that compared to NSP cells from the same patient, the SP subset consistently and significantly overexpressed genes associated with cellular activation, proliferation and survival including eIF4A1 (p&lt;.05), Wnt10a (p&lt;.05), Bcl2L12 (p&lt;.05) and Rel A (p=.07), a difference confirmed by qPCR. Conversely, NSP significantly overexpressed GSK3β (p&lt;.05) compared to SP. CD40 mediated activation of B-CLL tumor cells further enhanced relative expression of these genes in the SP fraction, and induced overexpression of the proliferation-associated genes Ki-67 and PCNA in SP compared to NSP cells by 8-fold and 132-fold, respectively. We found SP cells were more resistant to exposure to chemotherapeutic agents including 50 mM fludarabine than NSP. Analysis of results in patients who had received an autologous B-CLL cell tumor vaccine expressing CD40L and IL-2, however, showed that B-CLL SP cells were vulnerable to a T cell mediated immune response. Hence, 3–8 subcutaneous injections CD40L/IL-2 B-CLL cells had no immediate effect on total B-CLL cell numbers in the peripheral blood, but Hoechst analysis showed a disappearance of SP cells in 7/8 immunized patients, associated with a rise in B-CLL-reactive T cells. CD8+ T cell clones isolated and expanded with CD40-activated autologous B-CLL tumor cells specifically responded to autologous SP cells, suggesting that SP depletion is mediated vaccine induced B-CLL-specific T cells. In 6/7 patients SP cells returned within a few weeks of completing the course of vaccination, coincident with a decline in T cell immunity. In one patient, however, immunity was longer lived and in this patient control of SP B-CLL cells persisted for more that 12 months. In this individual there was a delayed (by 6 months), but progressive disappearance of the bulk (non-SP) tumor cells. These results are consistent with the existence of an SP tumor precursor population of B-CLL, that can be targeted by immune T cells in vivo, and removal of which leads to later disappearance of their non-SP progeny.


2021 ◽  
Vol 13 (587) ◽  
pp. eaaz6314
Author(s):  
Xia Liu ◽  
Celine L. Hartman ◽  
Lingyun Li ◽  
Carolyn J. Albert ◽  
Fusheng Si ◽  
...  

The functional state of T cells is a key determinant for effective antitumor immunity and immunotherapy. Cellular metabolism, including lipid metabolism, controls T cell differentiation, survival, and effector functions. Here, we report that development of T cell senescence driven by both malignant tumor cells and regulatory T cells is a general feature in cancers. Senescent T cells have active glucose metabolism but exhibit unbalanced lipid metabolism. This unbalanced lipid metabolism results in changes of expression of lipid metabolic enzymes, which, in turn, alters lipid species and accumulation of lipid droplets in T cells. Tumor cells and Treg cells drove elevated expression of group IVA phospholipase A2, which, in turn, was responsible for the altered lipid metabolism and senescence induction observed in T cells. Mitogen-activated protein kinase signaling and signal transducer and activator of transcription signaling coordinately control lipid metabolism and group IVA phospholipase A2 activity in responder T cells during T cell senescence. Inhibition of group IVA phospholipase A2 reprogrammed effector T cell lipid metabolism, prevented T cell senescence in vitro, and enhanced antitumor immunity and immunotherapy efficacy in mouse models of melanoma and breast cancer in vivo. Together, these findings identify mechanistic links between T cell senescence and regulation of lipid metabolism in the tumor microenvironment and provide a new target for tumor immunotherapy.


Blood ◽  
2007 ◽  
Vol 110 (1) ◽  
pp. 450-460 ◽  
Author(s):  
James N. Kochenderfer ◽  
Jessica L. Simpson ◽  
Christopher D. Chien ◽  
Ronald E. Gress

Development of CD8+ T-cell responses targeting tumor-associated antigens after autologous stem cell transplantations (ASCTs) might eradicate residual tumor cells and decrease relapse rates. Because thymic function dramatically decreases with aging, T-cell reconstitution in the first year after ASCT in middle-aged patients occurs primarily by homeostatic peripheral expansion (HPE) of mature T cells. To study antigen-specific T-cell responses during HPE, we performed syngeneic bone marrow transplantations (BMTs) on thymectomized mice and then vaccinated the mice with peptides plus CpG-containing oligodeoxynucleotides (CpGs) in incomplete Freund adjuvant and treated the mice with systemic interleukin-2 (IL-2). When CD8+ T-cell responses were measured ex vivo, up to 9.1% of CD8+ T cells were specific for tumor-associated epitopes. These large T-cell responses were generated by synergism between CpG and IL-2. When we injected mice subcutaneously with tumor cells 14 days after BMT and then treated them with peptide + CpG-containing vaccines plus systemic IL-2, survival was increased and tumor growth was inhibited in an epitope-specific manner. Depletion of CD8+ T cells eliminated epitope-specific antitumor immunity. This is the first report to demonstrate that CD8+ T-cell responses capable of executing antitumor immunity can be elicited by CpG-containing vaccines during HPE.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Cheng-Tao Jiang ◽  
Kai-Ge Chen ◽  
An Liu ◽  
Hua Huang ◽  
Ya-Nan Fan ◽  
...  

AbstractModulating effector immune cells via monoclonal antibodies (mAbs) and facilitating the co-engagement of T cells and tumor cells via chimeric antigen receptor- T cells or bispecific T cell-engaging antibodies are two typical cancer immunotherapy approaches. We speculated that immobilizing two types of mAbs against effector cells and tumor cells on a single nanoparticle could integrate the functions of these two approaches, as the engineered formulation (immunomodulating nano-adaptor, imNA) could potentially associate with both cells and bridge them together like an ‘adaptor’ while maintaining the immunomodulatory properties of the parental mAbs. However, existing mAbs-immobilization strategies mainly rely on a chemical reaction, a process that is rough and difficult to control. Here, we build up a versatile antibody immobilization platform by conjugating anti-IgG (Fc specific) antibody (αFc) onto the nanoparticle surface (αFc-NP), and confirm that αFc-NP could conveniently and efficiently immobilize two types of mAbs through Fc-specific noncovalent interactions to form imNAs. Finally, we validate the superiority of imNAs over the mixture of parental mAbs in T cell-, natural killer cell- and macrophage-mediated antitumor immune responses in multiple murine tumor models.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A120-A120
Author(s):  
Sashi Kasimsetty ◽  
Himavanth Gatla ◽  
Dhana Chinnasamy

BackgroundMCY-M11, an anti-mesothelin CAR (Meso-CAR) mRNA transfected PBMC cell product manufactured through <1 day-process is under clinical evaluation for the treatment of advanced ovarian cancer and peritoneal mesothelioma. In this in-vitro study, we characterized the phenotypic and functional status of immune cell populations in MCY-M11 and their possible role in antitumor immunity.MethodsMCY-M11 cell product were generated using unmanipulated healthy donor PBMCs (n=5) by transfection of Meso-CAR mRNA using MaxCyte’s proprietary Flow Electroporation® system. Frozen MCY-M11 cell product was thawed and cultured for 18 hours, then co-cultured with hMSLNneg or hMSLNpos human mesothelioma cell line, MSTO-211H, or stimulated with anti-CD3/anti-CD28 antibodies in vitro for 8 days. Distinct cell populations in MCY-M11 were evaluated for kinetics and duration of CAR expression, differentiation, activation, exhaustion, and their ability to secrete various immunomodulatory molecules during in vitro stimulation. Antigen-specific proliferation and cytotoxicity of MCY-M11 against hMSLNpos tumor cells as well as their ability to mount long-term antitumor immunity through epitope spreading mechanisms were studied.ResultsIndividual cell populations in MCY-M11 exhibited a consistent but transient Meso-CAR expression persisting for about 7 days. Cell subsets in MCY-M11 acquired early signs of activation and differentiation within 18–24 hours post-culture, but only attained full activation and lineage-specific differentiation upon specific response to hMSLNpos tumor cells. hMSLN antigen experienced MCY-M11 retained significant fractions of Naïve and Central Memory T cells and increased percentage of Effector Memory T cells along with increased expression of CD62L, CD27, and chemokine receptors (CCR5, CCR7, and CXCR3). MCY-M11 exhibited strong antigen-specific cytotoxicity against hMSLNpos tumor cells with corresponding increase in activation and proliferation of CD4+ and CD8+ T cell subsets and displayed low or no acquisition of known exhaustion markers. NK cells also exhibited a functionally superior molecular signature exhibiting increased levels of NKG2D, NKp44, NKp46, FAS, and TRAIL. The Monocytes and B cells in MCY-M11 also acquired an activated, differentiated, and mature phenotype, expressing molecules required for antigen presentation (HLA-DR, HLA-ABC, and CD205) and T cell co-stimulation (CD80 and CD86) to mount a strong antitumor response. These phenotypic changes in cell subsets of MCY-M11 transpired with simultaneous secretion of potent immunostimulatory molecules and chemokines facilitating an extended antitumor response through epitope spreading.ConclusionsWe demonstrated that MCY-M11 is a unique cell product possessing a complete built-in immune cellular machinery with favorable phenotype and enhanced functions specialized in mediating an effective and long-term antitumor response.Trial RegistrationNCT03608618


Antibodies ◽  
2021 ◽  
Vol 10 (3) ◽  
pp. 25
Author(s):  
Violet Y. Tu ◽  
Asma Ayari ◽  
Roddy S. O’Connor

T cell therapies, including CAR T cells, have proven more effective in hematologic malignancies than solid tumors, where the local metabolic environment is distinctly immunosuppressive. In particular, the acidic and hypoxic features of the tumor microenvironment (TME) present a unique challenge for T cells. Local metabolism is an important consideration for activated T cells as they undergo bursts of migration, proliferation and differentiation in hostile soil. Tumor cells and activated T cells both produce lactic acid at high rates. The role of lactic acid in T cell biology is complex, as lactate is an often-neglected carbon source that can fuel TCA anaplerosis. Circulating lactate is also an important means to regulate redox balance. In hypoxic tumors, lactate is immune-suppressive. Here, we discuss how intrinsic- (T cells) as well as extrinsic (tumor cells and micro-environmental)-derived metabolic factors, including lactate, suppress the ability of antigen-specific T cells to eradicate tumors. Finally, we introduce recent discoveries that target the TME in order to potentiate T cell-based therapies against cancer.


Sign in / Sign up

Export Citation Format

Share Document